Ethods IN Olecular Iology
Ethods IN Olecular Iology
Series Editor
John M. Walker
School of Life Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Stephen D. Skaper
Department of Pharmacology and Anesthesiology, University of Padova, Padova, Italy
Editor
Stephen D. Skaper, Ph.D.
Department of Pharmacology and Anesthesiology
University of Padova
Padova, Italy
[email protected]
Neuroscience has been described by Nobel laureate and neurobiologist Eric Kandel as the
“last frontier” of science. Nervous system development evolves from the well-orchestrated
processes of neural induction, cell proliferation, differentiation, cell migration, survival, and
synapse formation. Among these environmental cues, neurotrophic factors are secreted
proteins that promote neurite outgrowth, neuronal cell differentiation and survival both
in vivo and in vitro. Nerve growth factor (NGF) is the founding and best-characterized
member of the neurotrophin family of neurotrophic polypeptides and was discovered more
than half a century ago. Since their initial discovery, neurotrophic factors have raised expec-
tations that their clinical application to neurodegenerative diseases might provide an effec-
tive therapy for what are now untreatable conditions.
Exploring nervous system function and dysfunction is oftentimes impractical in humans,
and the availability of ex vivo and in vivo models which mimic, as closely as possible, how
neural cells act and interact among themselves is of critical importance in neurobiological
research. This volume of Methods in Molecular Biology aims to provide the reader, special-
ist and novice alike, with a selection of protocols and procedures which make use of cellular,
tissue, and whole animal models which can be applied to the investigation of neurotrophic
factors and other agents impacting on these systems. The book begins with a number of
chapters dealing with the culture of neurons and glia from the central and peripheral ner-
vous systems, neuron–glia coculture models, and cell-based assays for the evaluation of
neuroprotective molecules, as well as assays which can be applied to the study of agents
with neuroregenerative potential. Protocols describing viral- and nanoparticle-based deliv-
ery methods to neural cells are also presented, followed by chapters dealing with organo-
typic slice culture protocols. Lastly, several chapters are dedicated to in vivo lesion models
of relevance to nervous system pathology, which can be applied to the investigation of neu-
rotrophic factors and peptides.
I would like to gratefully acknowledge the contributors for their excellent cooperation
and patience during the course of this project. While extensive, this volume is by no means
intended to be all-inclusive, given the field’s vastness and publication space limitations.
Even so, I sincerely hope that this book will be useful to a broad spectrum of readers as they
explore nervous system physiology and pathology.
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 417
Contributors
xi
xii Contributors
Abstract
The neurotrophins are a family of closely related proteins that were first identified as survival factors for
sympathetic and sensory neurons and have since been shown to control a number of aspects of survival,
development, and function of neurons in both the central and peripheral nervous systems. Limiting quan-
tities of neurotrophins during development control the numbers of surviving neurons to ensure a match
between neurons and the requirement for a suitable density of target innervation. Biological effects of each
of the four mammalian neurotrophins are mediated through activation of one or more of the three mem-
bers of the tropomyosin-related kinase (Trk) family of receptor tyrosine kinases (TrkA, TrkB, and TrkC).
In addition, all neurotrophins activate the p75 neurotrophin receptor, a member of the tumor necrosis
factor receptor superfamily. Neurotrophin engagement of Trk receptors leads to activation of Ras, phos-
phatidylinositol 3-kinase, phospholipase C-γ1, and signaling pathways controlled through these proteins,
including the mitogen-activated protein kinases. Neurotrophin availability is required into adulthood,
where they control synaptic function and plasticity and sustain neuronal cell survival, morphology, and
differentiation. This chapter will provide an overview of neurotrophin biology, their receptors, and signal-
ing pathways.
Key words: Neurotrophic factors, Nerve growth factor, Brain-derived neurotrophic factor,
Neurotrophin-3, Neurotrophin-4/5, Glial cell line–derived neurotrophic factor, Tropomyosin-related
kinase, Receptor tyrosine kinases, Neurodegeneration, Neuroregeneration
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_1, © Springer Science+Business Media, LLC 2012
1
2 S.D. Skaper
Fig. 1. Target-derived, autocrine, and paracrine modes of neurotrophin presentation. During the period of target innervation,
neurotrophins support the survival of a restricted number of neurons expressing the appropriate tropomyosin-related
kinase (Trk) receptors. Limiting amounts of secreted neurotrophins do not allow for the survival of all neurons, which can,
however, be rescued by the administration of exogenous neurotrophins.
1 The Neurotrophin Family of Neurotrophic Factors: An Overview 3
2. Neurotrophin
Biology
The generality of the phenomenon of programmed cell death after
target deprivation (axotomy) has suggested that most neurons
respond to and are regulated by neurotrophic factors (9). This
hypothesis was validated by the subsequent isolation from pig brain
of a second neurotrophic factor, designated brain-derived neu-
rotrophic factor (BDNF) (10). Molecular cloning of the BDNF
gene (11) revealed its structural similarity to NGF, leading to the
concept of the neurotrophin family. Two additional members,
neurotrophin-3 (NT-3) and neurotrophin-4/5 (NT-4/5), were
later identified (12). The term neurotrophin-4/5 resulted from
uncertainties about whether the human neurotrophin-5 (13) was a
species homolog of the NT-4 found in Xenopus (14). The nomen-
clature NT-4/5 was subsequently adopted by a number of investi-
gators to denote the mammalian form of Xenopus NT-4 when it
was shown that the differences between the two factors were
only due to phylogenetic variation (15). Two novel neurotrophins
from the platyfish and carp have been cloned and designated
neurotrophin-6 (16) and neurotrophin-7 (17), respectively. These
do not have orthologs in mammals or birds and appear to interact
with the same receptors as the mammalian proteins. The neu-
rotrophins exhibit actions on distinct, as well as partially overlap-
ping, subsets of peripheral and central neurons (12, 18). Individual
neurons may also be responsive to more than one neurotrophin at
a given time or at subsequent times during development.
The mature neurotrophin proteins are noncovalently associ-
ated homodimers. The neurotrophins share a highly homologous
structure and are members of a large superfamily of growth factors
that contain a tertiary fold and cysteine “knot.” These features are
present in transforming growth factor-β, platelet-derived growth
factor, human chorionic gonadotropin, vascular endothelial growth
factor, and others. The cysteine knot consists of three disulfide
bonds that form a true knot of the polypeptide chain. Two cysteines
that make up the knot are missing from human neurotrophin-6.
Neurotrophin residues are generally divided into two categories,
conserved or variable, based on sequence alignments (12). Amino
acid residues implicated in neurotrophin binding that are conserved
are likely to represent a common interface to the tropomyosin-related
kinase (Trk) receptors, while the unique ones may represent elements
of specificity (19). The dimer interface is composed of β-strands
that maintain the conformation; these hydrophobic core residues
are highly conserved (20–22). In contrast to the β-strands, the
β-loops are highly variable. Detailed discussions of neurotrophin
structure and molecular evolution have been published (23–26).
4 S.D. Skaper
3. Neurotrophin
Receptors
The neurotrophins interact with two entirely distinct classes of
receptors, Trks and p75NTR, the first discovered member of the
tumor necrosis factor receptor superfamily. The former was initially
identified as a low-affinity receptor for NGF (32) but was subse-
quently shown to bind each of the neurotrophins with approxi-
mately equal nanomolar affinity (14, 33, 34). While p75
neurotrophin receptor (p75NTR) does not contain a catalytic motif,
it interacts with several proteins that relay signals important for
regulating neuronal cell survival, differentiation, and synaptic plas-
ticity. Each of the four cysteine-rich repeats of p75NTR participates
in binding to NGF (35). p75NTR binds NGF along the interface
between two NGF monomers, and binding results in a conforma-
tional change in NGF that alters the monomeric interface on the
opposite side of the NGF dimer, eliminating the potential for bind-
ing of one NGF dimer to two p75NTR monomers.
In mammals, the Trk subfamily of receptor tyrosine kinases
constitutes the second major class of neurotrophin receptors. The
extracellular domain of each of the Trks consists of a cysteine-rich
cluster followed by three leucine-rich repeats, another cysteine-rich
cluster, and two Ig-like domains (see Fig. 2). Each receptor has a
single transmembrane region that terminates in a cytoplasmic,
tyrosine kinase-containing domain surrounded by several tyrosine
residues that serve as phosphorylation-dependent docking sites for
cytoplasmic adaptors and enzymes. The neurotrophins dimerize
their cognate Trk receptor, resulting in activation via transphos-
phorylation of the cytoplasmic domain kinases. Specificity of neu-
rotrophin action is believed to be achieved in part by the selective
interaction between members of the Trk family of receptors and
the different neurotrophins. Thus, NGF binds to TrkA (36, 37),
TrkB binds BDNF and NT-4/5 with high affinity (38, 39), and
TrkC binds NT-3 (40). NT-3 can also interact, albeit with less effi-
ciency, with TrkA and TrkB (see Fig. 2) (15, 39).
Trk receptor function is modulated by p75NTR on several levels—
by promoting ligand binding, by promoting accessibility to neurotro-
phins through promotion of axonal growth and target innervation,
1 The Neurotrophin Family of Neurotrophic Factors: An Overview 5
Fig. 2. Neurotrophins and their receptors. The neurotrophins display specific interactions with the three Trk receptors: NGF
binds TrkA, BDNF and NT-4 bind TrkB, and NT-3 binds TrkC. In some cellular contexts, NT-3 can also activate TrkA and TrkB,
albeit with less efficiency. All neurotrophins bind to and activate p75 neurotrophin receptor (p75NTR). CR1–CR4 cysteine-rich
motifs, C1/C2 cysteine-rich clusters, LRR1–3 leucine-rich repeats, Ig1/Ig2 immunoglobulin-like domains.
the Trk receptors that account for the specificity observed in their
interactions (54). Such information may prove useful in the design
of small molecule mimetic ligands for neurotrophin receptors.
4. Neurotrophin
Signaling
Many extracellular signals transduce their cellular responses by reg-
ulating tyrosine phosphorylation of their target proteins. Ligand-
induced oligomerization of receptor protein tyrosine kinases and
autophosphorylation have been established as a general mechanism
for the activation of growth factor receptors, as well as many other
families of cell surface receptors (55). The Trk receptors are typical
receptor tyrosine kinases whose activation is stimulated by neu-
rotrophin-mediated dimerization and transphosphorylation of
activation loop kinases (56). Trk receptors are activated specifically
by the mature and not the pro-forms of the neurotrophin gene
products (57). Thus, the proteases that control processing of
proneurotrophins control Trk receptor responsiveness. The cyto-
plasmic domains of the Trk receptors contain several additional
tyrosines that are also substrates for phosphorylation by each recep-
tor’s tyrosine kinase. When phosphorylated, these residues form
the cores of binding sites that serve as a scaffolding for the recruit-
ment of a variety of adaptor proteins and enzymes that ultimately
propagate the neurotrophin signal (58). Within the activated Trk
molecule, the phosphotyrosines and their surrounding amino acid
residues create binding sites for proteins containing phosphoty-
rosine-binding or Src homology 2 domains. The major pathways
activated by the Trk receptors are Ras, Rac, phosphatidylinositol
3-kinase, phospholipase C-γ1, and their downstream effectors
(56, 58). In addition, endocytosis and transfer of Trk receptors to
different membrane compartments control the efficiency and dura-
tion of Trk-mediated signaling in part because many adaptor
proteins are localized to specific membrane compartments (59).
Activation of p75NTR results in activation of the nuclear factor-κB
and Jun kinase, as well as other signaling pathways (see Fig. 3).
Transactivation of receptor tyrosine kinases by G protein–cou-
pled receptors (GPCRs) is now recognized as an important signal-
ing mechanism allowing the cell to respond to a vast array of
extracellular stimuli (60, 61). Activation of TrkA and TrkB recep-
tors can also occur via a GPCR mechanism, in the absence of NGF
or BDNF (62, 63). Two GPCR ligands, adenosine and pituitary
adenylate cyclase–activating peptide, can activate Trk receptor
activity to increase the survival of neuronal cells through stimula-
tion of protein kinase B (Akt) activity. The effects of adenosine and
pituitary adenylate cyclase–activating peptide can be blocked by
K252a, an inhibitor of Trk tyrosine kinases. In contrast to
1 The Neurotrophin Family of Neurotrophic Factors: An Overview 7
Fig. 3. Neurotrophin signaling. Depicted are interactions of NGF (exemplar neurotrophin) with Trk and major intracellular
signaling pathways activated. Neurotrophin binding to Trk receptor leads to dimerization and autophosphorylation. The
linker Shc binds to phospho-Y490 on Trk and to a Grb2-SOS complex. SOS is a nucleotide exchange factor that activates
Ras by replacing GDP with GTP. Activated Ras interacts directly with the serine–threonine kinase Raf. The activated Raf
leads to the sequential activation of MAPK kinase (MEK), the mitogen-activated protein kinase-ERK kinase (MAPK). MAPK
translocates to the nucleus, where it phosphorylates transcription factors, promoting neuronal cell differentiation. Activation
of phosphatidylinositol 3-kinase through Ras or Gab1 promotes survival and growth of neurons. Activation of phospholi-
pase C-γ1 (PLC-γ1) results in activation of Ca2+- and protein kinase C-regulated pathways that promote synaptic
plasticity.
5. Why Study
Neurotrophins?
The power of neurotrophic factors to regulate neuronal cell sur-
vival in the developing nervous system and to promote also survival
after injury or protect neurons in toxin-mediated disease models in
8 S.D. Skaper
References
1. Levi-Montalcini R (1987) The nerve growth expressed in Xenopus ovary. Neuron 6,
factor 35 years later. Science 237, 1154–1162 845–858
2. Hamburger V, and Levi-Montalcini R (1949) 15. Ip NY, Stitt TN, Tapley P, Klein R, Glass DJ,
Proliferation, differentiation and degeneration Fandl J, et al. (1993) Similarities and differ-
in the spinal ganglia of the chick embryo under ences in the way neurotrophins interact with
normal and experimental conditions. J Exp the Trk receptors in neuronal and nonneuronal
Zool 111, 457–501 cells. Neuron 10, 137–149
3. Dreyfus CF (1989) Effects of nerve growth 16. Götz R, Köster R, Winkler C, Raulf F,
factor on cholinergic brain neurons. Trends Lottspeich F, Schartl M, et al. (1994)
Pharmacol Sci 10, 145–149 Neurotrophin-6 is a new member of the nerve
4. Thoenen H (1995) Neurotrophins and neu- growth factor family. Nature 372, 266–269
ronal plasticity. Science 270, 593–598 17. Lai KO, Fu WY, Ip FCF, and Ip NY (1998)
5. Ginty DD, and Segal RA (2002) Retrograde Cloning and expression of a novel neurotro-
neurotrophin signaling: Trk-ing along the phin, NT-7, from carp. Mol Cell Neurosci 11,
axon. Curr Opin Neurobiol 12, 268–274 64–76
6. Barde Y-A (1989) Trophic factors and neuronal 18. Eide FF, Lowenstein DH, and Reichardt LF
survival. Neuron 2, 1525–1534 (1993) Neurotrophins and their receptors-
7. Bothwell M (1995) Functional interactions of current concepts and implications for neuro-
neurotrophins and neurotrophin receptors. logic disease. Exp Neurol 121, 200–214
Annu Rev Neurosci 18, 223–253 19. Wiesmann C, Ultsch MH, Bass SH, and de Vos
8. Skaper SD (2008) The biology of neurotro- AM (1999) Crystal structure of nerve growth
phins, signalling pathways, and functional pep- factor in complex with the ligand-binding domain
tide mimetics of neurotrophins and their of the TrkA receptor. Nature 401, 184–188
receptors. CNS & Neurol Disorders – Drug 20. McDonald NQ, Lapatto R, Murray-Rust J,
Targets 7, 46–62 Gunning J, Wlodawer A, and Blundell TL
9. Oppenheim RW (1991) Cell death during (1991) New protein fold revealed by a 2.3-A
development of the nervous system. Annu Rev resolution crystal structure of nerve growth
Neurosci 14, 453–501 factor. Nature 354, 411–414
10. Barde Y-A, Edgar D, and Thoenen H (1982) 21. Robinson RC, Radziejewski C, Stuart DI, and
Purification of a new neurotrophic factor from Jones EY (1995) Structure of the brain-derived
mammalian brain. EMBO J 1, 549–553 neurotrophic factor/neurotrophin 3 heterodi-
11. Leibrock J, Lottspeich F, Hohn A, Hofer M, mer. Biochemistry 34, 4139–4146
Hengerer B, Masiakowski P, et al (1994) 22. Robinson RC, Radziejewski C, Spraggon G,
Molecular cloning and expression of brain- Greenwald J, Kostura MR, Burtnick LD, et al
derived neurotrophic factor. Nature 341, (1999) The structures of the neurotrophin 4
149–152 homodimer and the brain-derived neurotrophic
12. Ibáñez CF (1995) Neurotrophic factors: from factor/neurotrophin 4 heterodimer reveal a
structure-function studies to designing effec- common Trk-binding site. Protein Sci 8,
tive therapeutics. Trends Biotech 13, 217–227 2589–2597
13. Berkemeier LR, Winslow JW, Kaplan DR, 23. Lindsay RM, and Yancopoulos GD (1996)
Nicolics K, Goeddel DV, and Rosenthal A GDNF in a bind with known orphan: accessory
(1991) Neurotrophin-5: a novel neurotrophic implicated in new twist. Neuron 17, 571–574
factor that activates trk and trkB. Neuron 7, 24. Barde Y-A (1994) Neurotrophic factors: an
857–866 evolutionary perspective. J Neurobiol 25,
14. Hallböök F, Ibáñez CF, and Persson H (1991) 1329–1333
Evolutionary studies of the nerve growth factor 25. Kullander K, Carlson B, and Hallböök FJ
family reveal a novel member abundantly (1997) Molecular phylogeny and evolution of
10 S.D. Skaper
the neurotrophins from monotremes and mar- tyrosine protein kinase is a receptor for brain-
supials. Mol Evol 45, 311–321 derived neurotrophic factor and neurotro-
26. Butte MJ (2001) Neurotrophic factor struc- phin-3. Cell 66, 395–403
tures reveal clues to evolution, binding, speci- 39. Squinto SP, Stitt TN, Aldrich TH, Davis S,
ficity, and receptor activation. Cell Mol Life Sci Bianco SM, Radziejewski C, et al (1991) trkB
58, 1003–1013 encodes a functional receptor for brain-derived
27. Ip NY, and Yancopoulos GD (1996) The neu- neurotrophic factor and neurotrophin-3 but
rotrophins and CNTF: two families of collab- not nerve growth factor. Cell 65, 885–893
orative neurotrophic factors. Annu Rev 40. Lamballe F, Klein R, and Barbacid M (1991)
Neurosci 19, 491–415 trkC, a new member of the trk family of tyrosine
28. Doré S, Kar S, and Quirion R (1997) protein kinases, is a receptor for neurotro-
Rediscovering an old friend, IGF-I: potential phin-3. Cell 66, 967–979
use in the treatment of neurodegenerative dis- 41. Benedetti M, Levi A, and Chao MV (1993)
eases. Trends Neurosci 20, 326–331 Differential expression of nerve growth factor
29. Walicke P, Cowan WM, Ueno N, Baird A, and receptors leads to altered binding affinity and
Guillemin R (1986) Fibroblast growth factor neurotrophin responsiveness. Proc Natl Acad
promotes survival of dissociated hippocampal Sci USA 90, 7859–7863
neurons and enhances neurite extension. Proc 42. Bibel M, Hoppe E, and Barde Y-A (1999)
Natl Acad Sci USA 83, 3012–3016 Biochemical and functional interactions
30. Farkas LM, Dünker N, Roussa E, Unsicker K, between the neurotrophin receptors trk and
and Krieglstein K (2003) Transforming growth p75NTR. EMBO J 18, 616–622
factor-βs are essential for the development of 43. Mahadeo D, Kaplan L, Chao MV, and
midbrain dopaminergic neurons in vitro and Hempstead BL (1994) High affinity nerve
in vivo. J Neurosci 23, 5178–5186 growth factor binding displays a faster rate of
31. Miao N, Wang M, Ott JA, D’Alessandro JS, association than p140trk binding. J Biol Chem
Woolf TM, Bumcrot DA, et al (1997) Sonic 269, 6884–6891
hedgehog promotes the survival of specific 44. Esposito D, Patel P, Stephens RM, Perez P,
CNS neuron populations and protects these Chao MV, Kaplan DR, et al (2001) The cyto-
cells from toxic insult in vitro. J Neurosci 17, plasmic and transmembrane domains of the
5891–5899 p75 and Trk A receptors regulate high affinity
32. Radeke MJ, Misko TP, Hsu C, Herzenberg binding to nerve growth factor. J Biol Chem
LA, and Shooter E (1987) Gene transfer and 276, 32687–32695
molecular cloning of the rat nerve growth fac- 45. Curtis R, Adryan KM, Stark JL, Park JS,
tor receptor. Nature 325, 593–597 Compton DL, Weskamp G, et al (1995)
33. Rodriguez-Tébar A, Dechant G, and Barde Differential role of the low affinity neurotro-
Y-A (1990) Binding of brain-derived neu- phin receptor (p75) in retrograde axonal trans-
rotrophic factor to the nerve growth factor port of the neurotrophins. Neuron 14,
receptor. Neuron 4, 487–492 1201–1211
34. Rodríguez-Tébar A, Dechant G, Götz R, and 46. Makkerh JP, Ceni C, Auld DS, Vaillancourt F,
Barde Y-A (1992) Binding of neurotrophin-3 Dorval G, and Barker PA (2005) p75 neurotro-
to its neuronal receptors and interactions with phin receptor reduces ligand-induced Trk
nerve growth factor and brain-derived neu- receptor ubiquitination and delays Trk receptor
rotrophic factor. EMBO J 11, 917–922 internalization and degradation. EMBO J 6,
35. He XE, and Garcia KC (2004) Structure of 936–941
nerve growth factor complexed with the shared 47. Geetha T, Jiang J, and Wooten MW (2005)
neurotrophin receptor p75. Science 304, Lysine 63 polyubiquitination of the nerve
870–875 growth factor receptor TrkA directs internal-
36. Kaplan DR, Hempstead BL, Martin-Zanca D, ization and signaling. Mol Cell 20, 301–312
Chao MV, and Parada LF (1991) The trk 48. Bentley CA, and Lee KF (2000) p75 is impor-
proto-oncogene product: a signal transducing tant for axon growth and schwann cell migra-
receptor for nerve growth factor. Science 252, tion during development. J Neurosci 20,
554–558 7706–7715
37. Klein R, Jing SQ, Nanduri V, O’Rourke E, and 49. Harrison SMW, Jones ME, Uecker S, Albers
Barbacid M (1991) The trk proto-oncogene KM, Kudrycki KE, and Davis BM (2000)
encodes a receptor for nerve growth factor. Levels of nerve growth factor and neurotro-
Cell 65, 189–197 phin-3 are affected differentially by the pres-
38. Klein R, Nanduri V, Jing SA, Lambelle F, ence of p75 in sympathetic neurons in vivo.
Tapley P, Bryant S, et al (1991) The trkB J Comp Neurol 424, 99–110
1 The Neurotrophin Family of Neurotrophic Factors: An Overview 11
50. Lee K-F, Li E, Huber LJ, Landis SC, Sharpe 63. Lee FS, Rajagopal R, Kim AH, Chang PC, and
AH, Chao MV, et al (1992) Targeted mutation Chao MV (2002) Activation of Trk neurotro-
of the gene encoding the low affinity NGF phin receptor signaling by pituitary adenylate
receptor p75 leads to deficits in the peripheral cyclase-activating polypeptides. J Biol Chem
sensory nervous system. Cell 69, 737–749 277, 9096–9102
51. Lee KF, Bachman K, Landis S, and Jaenisch R 64. Rajagopal R, Chen ZY, Lee FS, and Chao MV
(1994) Dependence on p75 for innervation of (2004) Transactivation of Trk neurotrophin
some sympathetic targets. Science 263, receptors by G-protein-coupled receptor
1447–1449 ligands occurs on intracellular membranes.
52. Stucky CL, and Koltzenburg M (1997) The J Neurosci 24, 6650–6658
low-affinity neurotrophin receptor p75 regu- 65. Rajagopal R, and Chao MV (2006) A role for
lates the function but not the selective survival Fyn in Trk receptor transactivation by
of specific subpopulations of sensory neurons. J G-protein-coupled receptor signaling. Mol Cell
Neurosci 17, 4398–4405 Neurosci 33, 36–46
53. Ultsch MH, Wiesmann C, Simmons LC, 66. Sofroniew MV, Howe CL, and Mobley WC
Henrich J, Yang M, Reilly D, et al (1999) (2001) Nerve growth factor signaling, neuro-
Crystal structures of the neurotrophin-binding protection, and neural repair. Annu Rev
domain of TrkA, TrkB and TrkC. J Mol Biol Neurosci 24, 1217–1281
290, 149–159 67. Chao MV (2003) Neurotrophins and their
54. Urfer R, Tsoulfas P, O’Connell L, Hongo JA, receptors: a convergence point for many signal-
Zhao W, and Presta LG (1998) High resolu- ling pathways. Nat Rev Neurosci 4, 299–309
tion mapping of the binding site of TrkA for
68. Lu B, Pang PT, and Woo NH (2005) The yin
nerve growth factor and TrkC for neurotro-
and yang of neurotrophin action. Nat Rev
phin-3 on the second immunoglobulin-like
Neurosci 6, 603–614
domain of the Trk receptors. J Biol Chem 273,
5829–5840 69. Reichardt LF (2006) Neurotrophin-regulated
55. Weiss A, and Schlessinger J (1998) Switching signalling pathways. Phil Trans Royal Soc B
signals on or off by receptor dimerization. Cell 361, 1545–1564
94, 277–280 70. Hefti F (1986) Nerve growth factor promotes
56. Huang EJ, and Reichardt LF (2003) Trk recep- survival of septal cholinergic neurons after fim-
tors: roles in neuronal signal transduction. brial transactions. J Neurosci 6, 2155–2162
Annu Rev Biochem 72, 609–642 71. Williams LR, Varon S, Peterson GM, Wictorin
57. Lee R, Kermani P, Teng KK, and Hempstead K, Fischer W, Bjorklund A, et al (1986)
BL (2001) Regulation of cell survival by Continuous infusion of nerve growth factor
secreted proneurotrophins. Science 294, prevents basal forebrain neuronal death after
1945–1948 fimbria fornix transaction. Proc Natl Acad Sci
USA 83, 9231–9235
58. Segal RA, and Greenberg ME (1996)
Intracellular signaling pathways activated by 72. Kordower JH, Emborg ME, Bloch J, Ma SY,
neurotrophic factors. Annu Rev Neurosci 19, Chu Y, Leventhal L, et al (2000)
463–489 Neurodegeneration prevented by lentiviral vec-
59. York RD, Molliver DC, Grewal SS, Stenberg tor delivery of GDNF in primate models of
PE, McCleskey EW, Stork PJS (2000) Role of Parkinson’s disease. Science 290, 767–773
phosphoinositide 3-kinase and endocytosis in 73. Yasuda T, and Mochizuki H (2010) Use of
nerve growth factor-induced extracellular sig- growth factors for the treatment of Parkinson’s
nal-regulated kinase activation via Ras and disease. Expert Rev Neurother 10, 915–924
Rap1. Mol Cell Biol 20, 8069–8083 74. Podulso JF, Curran GL, and Gill JS (1998)
60. Daub H, Weiss FU, Wallasch C, and Ullrich A Putrescine-modified nerve growth factor: bio-
(1996) Role of transactivation of the EGF activity, plasma pharmacokinetics, blood-brain/
receptor in signalling by G-protein-coupled nerve barrier permeability, and nervous system
receptors. Nature 379, 557–560 biodistribution. J Neurochem 71, 1651–1660
61. Luttrell LM, Daaka Y, and Lefkowitz RJ (1999) 75. Wu D, and Pardridge WM (1999)
Regulation of tyrosine kinase cascades by Neuroprotection with noninvasive neurotro-
G-protein-coupled receptors. Curr Opin Cell phin delivery to the brain. Proc Natl Acad Sci
Biol 11, 177–180 USA 96, 254–259
62. Lee FS, and Chao MV (2001) Activation of 76. Zhang Y, and Pardridge WM (2001)
Trk neurotrophin receptors in the absence of Neuroprotection in transient focal brain isch-
neurotrophins. Proc Natl Acad Sci USA 98, emia after delayed intravenous administration
3555–3560 of brain-derived neurotrophic factor conjugated
12 S.D. Skaper
to a blood-brain barrier drug targeting system. 82. Garcia P, Youssef I, Utvik JK, Florent-Béchard
Stroke 32, 1378–1384 S, Barthélémy V, Malaplate-Armand C, et al
77. Choi-Lundberg DL, Lin Q, Chang YN, Chiang (2010) Ciliary neurotrophic factor cell-based
YL, Hay CM, Mohajeri H, et al (1997) delivery prevents synaptic impairment and
Dopaminergic neurons protected from degen- improves memory in mouse models of
eration by GDNF gene therapy. Science 275, Alzheimer’s disease. J Neurosci 30, 7516–7527
838–841 83. Brewster WJ, Fernyhough P, Diemel LT,
78. Zhou L, Baumgartner BJ, Hill-Felberg SJ, Mohiuddin L, and Tomlinson RR (1994)
McGowen LR, and Shine HD (2003) Diabetic neuropathy, nerve growth factor and
Neurotrophin-3 expressed in situ induces other neurotrophic factors. Trends Neurosci
axonal plasticity in the adult injured spinal cord. 17, 321–325
J Neurosci 23, 1424–1431 84. Apfel SC (2002) Nerve growth factor for the
79. Blesch A, Yang H, Weidner N, Hoang A, and treatment of diabetic neuropathy: what went
Otero D (2004) Axonal responses to cellularly wrong, what went right, and what does the future
delivered NT-4/5 after spinal cord injury. Mol hold? Internatl Rev Neurobiol 50, 393–413
Cell Neurosci 27, 190–201 85. Apfel SC, Kessler JA, Adornato BT, Litchy WJ,
80. Tuszynski MH, Thal L, Pay M, Salmon DP, U Sanders C, and Rask CA (1998) Recombinant
HS, Bakay R, et al (2005) A phase 1 clinical human nerve growth factor in the treatment of
trial of nerve growth factor gene therapy for diabetic polyneuropathy. NGF Study Group.
Alzheimer disease. Nat Med 11, 551–555 Neurology 51, 695–702
81. Qu HY, Zhang T, Li XL, Zhou JP, Zhao BQ, 86. McArthur JC, Yiannoutsos C, Simpson DM,
Li Q, et al (2008) Transducible P11-CNTF Adornato BT, Singer EJ, Hollander H, et al
rescues the learning and memory impairments (2000) A phase II trial of nerve growth factor
induced by amyloid-beta peptide in mice. Eur J for sensory neuropathy associated with HIV
Pharmacol 594, 93–100 infection. Neurology 54, 1080–1088
Chapter 2
Abstract
During development of the nervous system, neurons extend axons over considerable distances in a highly
stereospecific fashion in order to innervate their targets in an appropriate manner. This involves the recog-
nition, by the axonal growth cone, of guidance cues that determine the pathway taken by the axons. These
guidance cues can act to promote and/or repel growth cone advance. The directed growth of axons is
partly governed by cell adhesion molecules (CAMs) on the neuronal growth cone that bind to CAMs on
the surface of other axons or nonneuronal cells. In vitro assays have established the importance of the
CAMs ((neural cell adhesion molecule NCAM), N-cadherin, and L1) in promoting axonal growth over
cells. Compelling evidence implicates the fibroblast growth factor receptor tyrosine kinase as the primary
signal transduction molecule in the CAM pathway. CAMs are important constituents of synapses, and they
appear to play important and diverse roles in regulating synaptic plasticity associated with learning and
memory. Synthetic NCAM peptide mimetics corresponding to the binding site of NCAM for the fibro-
blast growth factor receptor promote synaptogenesis, enhance presynaptic function, and facilitate memory
consolidation. Dimeric versions of functional binding motifs of N-cadherin behave as N-cadherin agonists,
promoting both neuritogenesis and neuronal cell survival. Negative extracellular signals that physically
direct neurite growth have also been described. The latter include the myelin inhibitory proteins, Nogo,
myelin-associated glycoprotein, and oligodendrocyte-myelin glycoprotein. Potentiation of outgrowth-
promoting signals, together with antagonism of myelin proteins or their convergent receptor, NgR, and
its second messenger pathways, may provide new opportunities in the rational design of treatments for
acute brain injury and neurodegenerative disorders.
Key words: Cell adhesion molecules, Fibroblast growth factor receptor, Neuroprotection,
Neuroregeneration, Myelin, Nogo, Rho kinase
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_2, © Springer Science+Business Media, LLC 2012
13
14 S.D. Skaper
2. CAMs, Neuronal
Plasticity, and
Neuroprotection
Cell–cell interactions mediated by CAMs are fundamental to
numerous developmental processes. In the nervous system, the
ability of neurons to extend axons and innervate their targets in an
appropriate manner is governed to a large extent by the binding of
CAMs on the surface of other axons or nonneuronal cells (19, 20).
The neuronal receptors important for general cell contact–depen-
dent axonal growth are the β1-integrins, which recognize extracel-
lular matrix molecules; and in mammals, three CAMs, namely, the
neural cell adhesion molecule (NCAM), N-cadherin, and L1, pro-
mote axonal growth during development (2).
N-Cadherin is a member of the classical cadherin family of
transmembrane glycoproteins that mediate cellular recognition
via homophilic (binding with other N-cadherin molecules on
neighboring cells) interaction (21). In the nervous system, N-cadherin
function has been implicated in cell migration (22), axonal growth
and guidance (23), and synapse formation and synaptic plasticity
(24, 25). In addition to homophilic binding, cadherins have been
shown to interact with many adaptor or signaling molecules,
2 Neuronal Growth-Promoting and Inhibitory Cues in Neuroprotection… 15
Fig. 1. Strategies for central nervous system regeneration. Mechanical injury or traumatic injury, or chronic neurodegen-
erative disease, can result in neurons being “disconnected” from their innervation fields, leading to a loss of critical tissue-
derived trophic support. Regeneration may be promoted by replacement of neurotrophic factors, by stimulation of axonal
outgrowth and/or cell survival with adhesion molecules, and by agents capable of overriding the outgrowth inhibitory
environment. MAG myelin-associated glycoprotein, OMgp oligodendrocyte-myelin glycoprotein.
90
60
45
30
15
0
0 1 3 10 30 50
SW4 (ug/ml)
Fig. 2. The dimeric HAVDI peptide SW4 attenuates glutamate-induced toxicity in culture
hippocampal neurons. Cultures (days in vitro, 7–9) were pretreated with the indicated
concentrations of SW4 peptide for 48 h and then exposed to 50 μM glutamate, and neu-
ronal cell survival was quantified 24 h later by a colorimetric reaction (MTT). Data are
means ± SD (three experiments) expressed relative to no glutamate (100%). **P < 0.05 or
**
P < 0.01 vs. glutamate only. Reproduced with permission from Elsevier (75).
3. Growth
Inhibitory
Molecules
In contrast to fish, amphibia, and the mammalian peripheral nerves
and developing central nerves, adult central mammalian neurons
do not regrow functional axons after damage. It is not the absence
of growth-promoting molecules in the CNS, but rather the pres-
ence of axonal outgrowth inhibitors in CNS astroglial “scars” (50)
and CNS myelin (51, 52) that suppresses the regrowth of damaged
axons. In particular, two components present in CNS myelin have
been characterized as potent inhibitors of axonal growth: MAG
(16, 17) and Nogo-A, the largest transcript of the recently identi-
fied nogo gene (formerly called NI-220) (13–15). Other inhibitors
include chondroitin sulfate proteoglycans (53) and OMgp (18).
Nogo is a member of the Reticulon family and occurs in three
forms, Nogo-A, -B, and -C, which are generated from alternate
splicing (13–15). Nogo-A, -B, and -C all contain a 66-amino acid
ECD (Nogo-66) that alone can inhibit neurite outgrowth and
induce growth cone collapse (13–15). MAG is a type I membrane
protein composed of five extracellular immunoglobulin (Ig)–like
domains (16, 17), whereas OMgp is a glycosylphosphatidylinosi-
tol-anchored protein (18). Remarkably, a single neuronal protein,
the Nogo-66 receptor (NgR), binds Nogo, MAG, and OMgp
(54–57). The p75 low-affinity neurotrophin receptor protein has
been implicated in transducing a myelin/NgR signal to the axonal
interior (58, 59) (see Fig. 3).
The presence of myelin-derived inhibitors suggests that block-
ing their action might allow the intrinsic growth potential of CNS
axons to be unmasked (60). Indeed, neutralizing Nogo-A with
IN-1 antibody induced CNS axon regeneration and improved
recovery after various lesions (61, 62). Targeting the axonal NgR
with a competitive antagonist compound has the potential to block
the action of the three known myelin inhibitors. Both intrathecal
(63) and delayed systemic application (64) of NEP1–40 (Nogo
extracellular peptide, residues 1–40) produced significant axonal
regrowth after spinal cord hemitransection injury, as well as
enhanced locomotor recovery (64). However, these reagents only
target a single myelin protein, Nogo, which may not be sufficient to
18 S.D. Skaper
Oligodendrocyte C
NogoA MAG
?
? OMgp
Nogo-66
N
NgR
NgR3
NgR2
? p75 Neurite
outgrowth
inhibition
GPI GPI Axon PI
Rho A
ROCK Cytoskeleton
Fig. 3. The Nogo receptor and inhibition of axonal regeneration. The leucine-rich repeat
domains of the Nogo receptor (NgR) are necessary for interaction with Nogo-66, MAG, and
OMgp. NgR does not transduce signals directly but utilizes coreceptor molecules, for
example, p75 or others. Coreceptor activation, in turn, activates the Rho and ROCK path-
way to modulate the cytoskeleton and neurite growth. While structurally similar to NgR1,
NgR2 and NgR3 display essentially no binding to known myelin-derived NgR ligands. GPI
glycosylphosphatidylinositol, ROCK Rho-associated kinase.
4. Nerve
Regeneration
and Neurotrophic
Factors Information is encoded in the CNS through networks of neurons
that are functionally connected by synapses. Upon injury, factors
in CNS myelin inhibit neurite outgrowth. Understanding of how
signals from extracellular factors associated with myelin and the
injury site are integrated with outgrowth-promoting signals and
neurotrophic factors to regulate axonal elongation should further
facilitate the development of interventions to improve the outcome
of both acute CNS injury and chronic neurodegenerative
disorders. Methodologies which can be applied to understanding
how neurite stimulatory signals and neurotrophic factors interact
will be of considerable importance in advancing nerve regenera-
tion efforts. This volume describes as well protocols which have
this goal in mind.
20 S.D. Skaper
References
1. Tessier-Lavigne M, and Goodman CS (1996) 16. McKerracher L, David S, Jackson DL, Kottis V,
The molecular biology of axonal guidance. Dunn RJ, and Braun PE (1994) Identification
Science 274, 1123–1133 of myelin-associated glycoprotein as a major
2. Walsh FS, and Doherty P (1997) Neural cell myelin-derived inhibitor of neurite outgrowth.
adhesion molecules of the immunoglobulin Neuron 13, 805–811
superfamily: role in axonal growth and guid- 17. Mukhopadhyay G, Doherty P, Walsh FS,
ance. Annu Rev Cell Dev Biol 13, 425–456 Crocker PR, and Filbin MT (1994) A novel
3. Ip NY, and Yancopoulos GD (1996) The neu- role for myelin-associated glycoprotein as an
rotrophins and CNTF: two families of collab- inhibitor of axonal regeneration. Neuron 13,
orative neurotrophic factors. Annu Rev 757–767
Neurosci 19, 491–515 18. Wang KC, Koprivica V, Kim JA, Sivasankaran
4. Skaper SD, and Walsh FS (1998) Neurotrophic R, Guo Y, Neve RL, et al (2002)
molecules: strategies for designing effective Oligodendrocyte-myelin glycoprotein is a
therapeutic molecules in neurodegeneration. Nogo receptor ligand that inhibits neurite out-
Mol Cell Neurosci 12, 179–193 growth. Nature 417, 941–944
5. Serefini T, Kennedy TE, Galko MJ, Mirzayan 19. Dodd J, and Jessell JM (1988) Axon guidance
C, Jessell TM, and Tessier-Lavigne M (1994) and the patterning of neuronal projections in
The netrins define a family of axon outgrowth- vertebrates. Science 242, 692–699
promoting proteins homologous to C. elegans 20. Goodman CS, and Shatz CJ (1993)
UNC-6. Cell 78, 409–424 Developmental mechanisms that generate pre-
6. Mueller BK (1999) Growth cone guidance: cise patterns of neuronal connectivity. Cell/
first steps toward a deeper understanding. Annu Neuron 72(suppl. 10), 77–98
Rev Neurosci 22, 351–388 21. Takeichi M (1995) Morphogenetic role of clas-
7. Fournier AE, and Strittmatter SM (2001) sic cadherins. Curr Opin Cell Biol 7, 619–627
Repulsive factors and axon regeneration in the 22. Barami K, Kirschenbaum B, Lemmon V, and
CNS. Curr Opin Neurobiol 11, 89–94 Goldman SA (1994) N-Cadherin and
8. Kolodkin AL (1996) Semaphorins: mediators Ng-CAM/8D9 are involved serially in the
of repulsive growth cone guidance. Trends Cell migration of newly generated neurons into the
Biol 6: 15–22 adult songbird brain. Neuron 13, 567–582
9. Goodman CS (1996) Mechanisms and mole- 23. Matsunaga M, Hatta K, Nagafuchi A, and
cules that control growth cone guidance. Annu Takeichi M (1988) Guidance of optic nerve
Rev Neurosci 19, 341–377 fibers by N-cadherin adhesion molecules.
10. Kenwrick S, Watkins A, and De Angelis E Nature 334, 62–64
(2000) Neural cell recognition molecule L1: 24. Inoue A, and Sanes JR (1997) Lamina-specific
relating biological complexity to human disease connectivity in the brain: regulation by
mutations. Hum Mol Genet 9, 879–886 N-cadherin, neurotrophins, and glycoproteins.
11. Cajal RS (1928) Degeneration and Regeneration Science 276, 1428–1431
of the Nervous System. Hafner. New York. 25. Bozdagi O, Shan W, Tanaka H, Benson DL,
12. McGee AW, and Strittmatter SM (2003) The and Huntley GW (2000) Increasing numbers
Nogo-66 receptor: focusing myelin inhibition of synaptic puncta during late-phase LTP:
of axon regeneration. Trends Neurosci 26, N-cadherin is synthesized, recruited to synaptic
193–198 sites, and required for potentiation. Neuron
13. Chen MS, Huber AB, Van Der Haar ME, Frank 28, 245–259
M, Schnell L, Spillmann AA, et al (2000) 26. Saffell JL, Williams EJ, Mason IJ, Walsh FS,
Nogo-A is a myelin-associated neurite out- and Doherty P (1997) Expression of a domi-
growth inhibitor and an antigen for monoclo- nant negative FGF receptor inhibits axonal
nal antibody IN-1. Nature 403, 434–439 growth and FGF receptor phosphorylation
14. GrandPré T, Nakamura F, Vartanian T, and stimulated by CAMs. Neuron 18, 231–242
Strittmatter SM (2000) Identification of the 27. Williams E-J, Furness J, Walsh FS, and Doherty
Nogo inhibitor of axon regeneration as a P (1994) Activation of the FGF receptor
Reticulon protein. Nature 403, 439–444 underlies neurite outgrowth stimulated by L1,
15. Prinjha R, Moore SE, Vinson M, Blake S, N-CAM, and N-cadherin. Neuron 13,
Morrow R, Christie G, et al (2000) Inhibitor 583–594
of neurite outgrowth in humans. Nature 403, 28. Williams E-J, Williams G, Howell FV, Skaper
383–384 SD, Walsh FS, and Doherty P (2001)
2 Neuronal Growth-Promoting and Inhibitory Cues in Neuroprotection… 21
Identification of an N-cadherin motif that can 41. Benson DL, Schnapp LM, Shapiro L, and
interact with the fibroblast growth factor recep- Huntley GW (2000) Making memories stick:
tor and is required for axonal growth. J Biol cell adhesion molecules in synaptic plasticity.
Chem 276, 43879–43886 Trends Cell Biol 10, 473–482
29. Doherty P, Williams E, and Walsh FS (1995) A 42. Lüthi A, Laurent JP, Figurov A, Muller D, and
soluble form of the L1 glycoprotein stimulated Schachner M (1994) Hippocampal long-term
neurite outgrowth. Neuron 14, 57–66 potentiation and neural cell adhesion molecules
30. Utton MA, Eickholt B, Howell FV, Wallis J, L1 and NCAM. Nature 372, 777–779
and Doherty P (2001) Soluble N-cadherin 43. Rønn LCB, Bock E, Linnemann D, and Jahnsen
stimulates fibroblast growth factor receptor H (1995) NCAM-antibodies modulate induc-
dependent neurite outgrowth and N-cadherin tion of long-term potentiation in rat hippocam-
and the fibroblast growth factor receptor co- pal CA1. Brain Res 677, 145–151
cluster in cells. J Neurochem 76, 1421–1430 44. Doyle E, Nolan PM, Bell R, and Regan CM
31. Koch AW, Bozic D, Pertz O, and Engel J (1992) Intraventricular infusions of anti-neural
(1999) Homophilic adhesion by cadherins. cell adhesion molecules in discrete posttraining
Curr Opin Struct Biol 9, 275–281 period impair consolidation of a passive avoid-
32. Williams E, Williams G, Gour BJ, Blaschuk ance response in the rat. J Neurochem 59,
OW, and Doherty P (2000) A novel family of 1570–1573
cyclic peptide antagonists suggests that 45. Cremer H, Lange R, Christoph A, Plomann M,
N-cadherin specificity is determined by amino Vopper G, Roes J, et al (1994) Inactivation of
acids that flank the HAV motif. J Biol Chem the NCAM gene in mice results in size reduc-
275, 4007–4012 tion of the olfactory bulb and deficits in spatial
33. Williams EJ, Williams G, Gour B, Blaschuk O, learning. Nature 367, 455–459
and Doherty P (2000) INP, a novel N-cadherin 46. Rønn LCB, Olsen M, Ostergaard S, Kiselyov V,
antagonist targeted to the amino acids that Berezin V, Mortensen MT, et al (1999)
flank the HAV motif. Mol Cell Neurosci 15, Identification of a neuritogenic ligand of the
456–464 neural cell adhesion molecule using a combina-
34. Williams G, Williams E-J, and Doherty P torial library of synthetic peptides. Nat Biotech
(2002) Dimeric versions of two short 17, 1000–1005
N-cadherin binding motifs (HAVDI and 47. Saffell JL, Walsh FS, and Doherty P (1994)
INPISG) function as N-cadherin agonists. J Expression of NCAM containing VASE in
Biol Chem 277, 4361–4367 neurons can account for a developmental loss
35. Skaper SD, Facci, L, Williams G, Williams EJ, in their neuritic outgrowth response to NCAM
Walsh FS, and Doherty P (2004) A dimeric in a cellular substratum. J Cell Biol 125,
version of the short N-cadherin binding motif 427–436
HAVDI promotes neuronal cell survival by 48. Kiselyov VV, Skladchikova G, Hinsby AM,
activating an N-cadherin/fibroblast growth Jensen PH, Kulahin N, Soroka V, et al (2003)
factor receptor signalling cascade. Mol Cell Structural basis for a direct interaction between
Neurosci 26, 17–23 FGFR1 and NCAM and evidence for a regula-
36. Chen S, Mantei N, Dong L, and Schachner M tory role of ATP. Structure 11, 691–701
(1999) Prevention of neuronal cell death by 49. Cambon K, Hansen SM, Venero C, Herrero
neural cell adhesion molecules L1 and CHL1. J AI, Skibo G, Berezin V, et al (2004) A synthetic
Neurobiol 38, 428–439 neural cell adhesion molecule mimetic peptide
37. Ditlevsen DK, Køhler LB, Pedersen MV, Risell promotes synaptogenesis, enhances presynaptic
M, Kolkova K, Meyer M, et al (2003) The role function, and facilitates memory consolidation.
of phosphatidylinositol 3-kinase in neural cell J Neurosci 24, 4197–4204
adhesion molecule–mediated neuronal differen- 50. Fawcett JW, and Asher RA (1999) The glial
tiation and survival. J Neurochem 84, 546–556 scar and central nervous system repair. Brain
38. Goda Y (2002) Cadherins communicate struc- Res Bull 49, 377–391
tural plasticity of presynaptic and postsynaptic 51. Schwab ME, and Thoenen H (1985)
terminals. Neuron 35, 1–3 Dissociated neurons regenerate into sciatic, but
39. Togashi H, Abe K, Mizoguchi A, Takaoka K, not optic nerve explants irrespective of neu-
Chisaka O, and Takeichi M (2002) Cadherin rotrophic factors. J Neurosci 5, 2415–2423
regulates dendritic spine morphogenesis. 52. Schwab ME, and Caroni P (1988)
Neuron 35, 77–89 Oligodendrocytes and CNS myelin are nonper-
40. Schachner M (1997) Neural recognition mol- missive substrates for neurite growth and fibro-
ecules and synaptic plasticity. Curr Opin Cell blast spreading in vitro. J Neurosci 8,
Biol 9, 627–634 2381–2393
22 S.D. Skaper
53. Niederöst BP, Zimmermann DR, Schwab ME, recovery from spinal cord injury. J Neurosci
and Bandtlow CE (1999) Bovine CNS myelin 23, 4219–4227
contains neurite growth-inhibitory activity 65. Lee J-K, Kim J-E, Sivula M, and Strittmatter
associated with chondroitin sulphate proteo- SM (2004) Nogo receptor antagonism pro-
glycans. J Neurosci 19, 8979–8989 motes stroke recovery by enhancing axonal
54. Fournier AE, GrandPre T, and Strittmatter SM plasticity. J Neurosci 24, 6209–6217
(2001) Identification of a receptor mediating 66. Li W, Walus L, Rabacchi SA, Jirik A, Chang E,
Nogo-66 inhibition of axonal regeneration. Schauer J, (2004) A neutralizing anti-
Nature 409, 341–346 Nogo66 receptor monoclonal antibody
55. Domeniconi M, Cao Z, Spencer T, Sivasankaran reverses inhibition of neurite outgrowth by
R, Wang K, Nikulina E, et al (2002) Myelin- central nervous system myelin. J Biol Chem
associated glycoprotein interacts with the 279, 43780–43788
nogo66 receptor to inhibit neurite outgrowth. 67. Yu P, Huang L, Zou J, Yu Z, Wang Y, Wang Z,
Neuron 35, 283–290 et al (2008) Immunization with recombinant
56. Liu BP, Fournier A, GrandPré T, and Nogo-66 receptor (NgR) promotes axonal
Strittmatter SM (2002) Myelin-associated gly- regeneration and recovery of function after spinal
coprotein as a functional ligand for the Nogo- cord injury in rats. Neurobiol Dis 32, 535–542
66 receptor. Science 297, 1190–1193 68. Luo L (2000) Rho GTPases in neuronal mor-
57. Wang KC, Koprivica V, Kim JA, Sivasankaran phogenesis. Nat Rev Neurosci 1, 173–180
R, Guo Y, Neve RL, et al (2002) Oligodendrocyte- 69. Bito H, Furuyashiki T, Ishihara H, Shibasaki Y,
myelin glycoprotein is a Nogo receptor ligand Ohashi K, Mizuno K (2000) A critical role for
that inhibits neurite outgrowth. Nature 417, Rho-associated kinase, p160ROCK, in deter-
941–944 mining axonal outgrowth in mammalian CNS
58. Wang KC, Kim JA, Sivasankaran R, Segal R, neurons. Neuron 26, 431–441
and He Z (2002) p75 interacts with the Nogo 70. Yamashita T, Higuchi H, and Tohyama M
receptor as a co-receptor for Nogo, MAG, and (2002) The p75 receptor transduces the signal
OMgp. Nature 420, 74–78 from myelin-associated glycoprotein to Rho.
59. Wong ST, Henley JR, Kanning KC, Huang J Cell Biol 157, 565–570
KH, Bothwell M, and Poo MM (2002) A p75 71. Fournier AE, Takizawa BT, and Strittmatter
(NTR) and Nogo receptor complex mediates SM (2003) Rho kinase inhibition enhances
repulsive signaling by myelin-associated glyco- axonal regeneration in the injured CNS. J Neurosci
protein. Nat Neurosci 5, 1302–1308 23, 1416–1423
60. Lee DHS, Strittmatter SM, and Sah DWY 72. Niederöst B, Oertle T, Fritsche J, McKinney
(2003) Targeting the Nogo receptor to treat RA, and Bandtlow CE (2002) Nogo-A and
central nervous system injuries. Nat Rev Drug myelin-associated glycoprotein mediate neurite
Discovery 2, 872–879 growth inhibition by antagonistic regulation of
61. Bregman BS, Kunkel-Bagden E, Schnell L, Dai RhoA and Rac1. J Neurosci 22, 10368–10376
HN, Gao D, and Schwab ME (1995) Recovery 73. Dergham P, Ellezam B, Essagian C, Avedissian
from spinal cord injury mediated by antibodies H, Lubell WD, and McKerracher L (2002)
to neurite growth inhibitors. Nature 378, Rho signalling pathway targeted to promote
498–501 spinal cord repair. J Neurosci 22, 6570–6577
62. Thallmair M, Metz GA, Z’Graggen WJ, 74. Kubo T, Hata, K, Yamaguchi A, and Yamashita
Raineteau O, Kartje GL, and Schwab ME T (2007) Rho-ROCK inhibitors as emerging
(1998) Neurite growth inhibitors restrict plas- strategies to promote nerve regeneration. Curr
ticity and functional recovery following corti- Pharm Des 13, 2493–2499
cospinal tract lesions. Nat Neurosci 1, 75. Skaper, S.D., Facci, L., Gilliams, G., Williams,
124–131 E.-J., Walsh, F.S. and P. Doherty (2004) A
63. GrandPre T, Li S, and Strittmatter SM (2002) dimeric version of the short N-cadherin bind-
Nogo-66 receptor antagonist peptide promotes ing motif HAVDI promotes neuronal cell sur-
axonal regeneration. Nature 417, 547–551 vival by activating an N-cadherin/fibroblast
64. Li S, and Strittmatter SM (2003) Delayed sys- growth factor receptor signalling cascade. Mol
temic Nogo-66 receptor antagonist promotes and Cell Neurosc 26, 17–23
Chapter 3
Abstract
In primary culture of the early postnatal cerebellum, glutamatergic granule cells are highly enriched and
recapitulate many properties characteristic of developing granule neurons in vivo. For example, withdrawal
of K+ from differentiated rat primary cerebellar granule neurons results in the apoptotic death of the majority
of cells after 48 h. Removal of cerebellar granule neurons from depolarizing culture conditions with high
K+ is thought to reflect the regulation of trophic action of neuronal activity and has found widespread
application as a model for studying the mechanisms of survival factor withdrawal-induced neuronal cell
apoptosis and the neuroprotective action of trophic agents. This chapter presents a protocol for the culture
of postnatal rat cerebellar granule neurons and results in a preparation containing 95% glutamatergic granule
cells and its application to the evaluation of corticotropin receptor agonists as neuroprotective agents.
Key words: Cerebellum, Granule neurons, Cell culture, Rat, Depolarization, Development,
Phosphatidylinositol 3-kinase, Glycogen synthase kinase-3, Apoptosis, Neuroprotection,
Neurotrophic factors
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_3, © Springer Science+Business Media, LLC 2012
23
24 L. Facci and S.D. Skaper
Ser9/21 P
GSK-3 GSK-3
Less active
Fig. 1. Schematic representation of the phosphatidylinositol 3-kinase (PI3K)/glycogen synthase kinase (GSK-3) signaling
pathway involved in cerebellar granule neuron survival promoted by a high-potassium environment. Phosphorylation of
protein kinase B (PKB or Akt) on Ser473 leads to its activation, with subsequent phosphorylation (and inactivation) of GSK-3
on Ser9 and Ser21. The active state of GSK-3 promotes apoptosis, while GSK-3 inactivation is antiapoptotic. RTK receptor
tyrosine kinase; PDK1 phosphoinositide-dependent kinase-1.
3 Culture of Rat Cerebellar Granule Neurons and Application… 25
2. Materials
2.3. Culture Media 1. Krebs Buffer (10×). Add the following components and
and Other Solutions amounts: NaCl (17.67 g), KCl (0.900 g), KH2PO4 (0.415 g),
D-glucose (6.425 g), NaHCO3 (5.350 g), 0.5% phenol red
(5 mL), dissolve in water and bring to 250 mL, and filter-
sterilize. The solution can be stored at 4°C for 3 months.
2. Solution K. Dilute the 10× Krebs buffer stock 1:10 in double-
distilled water, add MgCl2 to 0.5 mM and 0.3% (w/v) BSA.
Filter-sterilize. Can be stored for 1 month at 4°C.
3. 0.15 M Borate buffer, pH 8.4. Dissolve 28.6 g of sodium borate
(Na2B4O7 ⋅ 10 H2O) in 500 mL water (pH will be ~9.2). Adjust
pH to 8.4 with 5 N HCl. Filter-sterilize and store at 4°C (up
to 6 months).
4. Ara-C stock solution (10 mM). Prepare Ara-C as 10-mM stock
solution in PBS, filter-sterilize, and store as aliquots at −20°C
for up to 6 months. Once thawed, the aliquot tube may be
kept at 4°C for 4 weeks (see Note 2).
5. Serum-free culture medium. This consists of Neurobasal-A
medium (see Note 3) with the addition of B27 supplements
(1:50 of a 50× stock solution), 1 mM sodium pyruvate (from
a 100-mM stock solution), 2 mM L-glutamine (from a 200-
mM stock solution), 20 mM KCl (1:50 from a 1-M stock
solution), 100 U/mL penicillin-100 μg/mL streptomycin
3 Culture of Rat Cerebellar Granule Neurons and Application… 27
2.5. Papain 1. The kit consists of the following components: vial (1)—100 mL
Dissociation System: EBSS (Earle’s balanced salt solution); vial (2)—papain con-
Solutions taining L-cysteine and EDTA; vial (3)—DNase; vial (4)—ovo-
mucoid protease inhibitor with BSA.
2. Add 2 mL of 1 M HEPES buffer to the entire contents of vial
(1)—this will still be referred to as “EBSS.”
3. Solution A: Add 5 mL of EBSS to vial (2) (20 U of papain/mL
in 1 mM L-cysteine with 0.5 mM EDTA).
4. Solution B: Add to vial (3) 0.5 mL of EBSS (2,000 U/mL
DNase).
5. Solution C: Add 32 mL of EBSS to vial (4) (10 mg ovomucoid
inhibitor and 10 mg BSA/mL). This solution is stable when
stored at 2–8°C.
6. Solution D: Add 250 μL of solution B to 5 mL of solution A.
28 L. Facci and S.D. Skaper
3. Methods
3.1. Tissue Dissection 1. Dissect out the cerebellum from 7- to 8-day-old rat pups of
and Dissociation both sexes (CD strain, Sprague Dawley) under aseptic condi-
(Papain Dissociation tions following decapitation. Pups are sacrificed in accordance
Procedure) with appropriate institutional and national guidelines for the
care and use of laboratory animals. Standard techniques and
anatomical landmarks are followed.
2. Make an incision under the skin along the midline of the dorsal
surface and peel back the skin. Make a second incision close to
the midline along the exposed dorsal surface (the cut should
be along the complete anteroposterior axis of the lateral ven-
tricles). Splay open the hemispheres along the cut surface and
continue cut along the dorsal surface of the hemisphere.
3. Remove the cerebellum to a 10-cm ∅ dish containing solution
K. Free the tissue of meninges with the use of a stereo dissect-
ing microscope.
4. Remove solution K and mince the tissue with a flamed razor
blade. Collect the tissue pieces with solution K and transfer to
a 15-mL centrifuge tube.
5. Allow the tissue pieces to settle by gravity, remove the medium,
and add 5 mL of solution D. Incubate for 15 min at 37°C with
occasional agitation.
6. Vigorously titrate with a cotton-plugged, sterile Pasteur pipette
(attached to an automatic pipettor) until devoid of clumps (see
Notes 7 and 8).
7. Centrifuge at 200 × g for 5 min.
8. During step 5, prepare two tubes with 2.7 mL EBSS + 0.3 mL
solution C + 0.15 mL solution B (3.15 mL total = EBSS Mix);
5 mL solution C.
9. After centrifugation, discard the supernatant and resuspend
the pellet in 3 mL EBSS Mix. Using a Pasteur pipette, carefully
layer the cell suspension on top of the 5 mL ovomucoid.
10. Centrifuge at 150 × g for 5 min.
3 Culture of Rat Cerebellar Granule Neurons and Application… 29
11. Remove and discard the supernatant and resuspend the pellet
in culture medium (either serum-containing or serum-free).
12. It will be necessary to dilute the cell suspension before counting.
A density of 1.5–2 million cells/mL for hemacytometer counting
is convenient. See Subheading 3.3 for details on counting with
the hemacytometer. Typical yields are 20–23 million cells/
cerebellum for 7–8-day-old animals.
13. Dilute the cell suspension with complete culture medium to
give the desired plated density. Generally, it is advisable to plate
2.5–3.0 × 105 cells/cm2 of culture surface area. Survival of
CGN is density-dependent, and plating the cells below 1 × 105
cells/cm2 will result in reduced cell viability (see Notes 9–11).
14. Add Ara-C 18–22 h after cell plating to a final concentration of
10 μM (1:1,000 from a 10-mM stock solution in PBS). Ara-C is
an antimitotic and is added to inhibit the growth of nonneurons.
3.2. Preparation 1. Collect cerebellar tissue from 7- to 8-day-old rat pups into
of CGNs Using Trypsin solution K, as described in Subheading 3.1. Mince tissue with
Dissociation a razor blade and collect in a 15-mL tube, then add solution B
(1 mL/cerebellum). If more than 10 cerebella are processed,
it is advisable to perform this step using a 50-mL tube.
2. Incubate for 15 min (37°C) with gentle swirling.
3. Add solution D (1 mL/cerebellum) to the tube and centrifuge
at 300 × g for 3 min.
4. Remove as much as possible the supernatant; the pellet may
not be very compact. It is possible to transfer the pellet to a
new tube, thereby eliminating most of the supernatant.
5. Add 2 mL of solution C. Pipette C down 5 times with a 5-mL
graduated pipette to break up the largest tissue pieces. Dissociate
the tissue using a long (9-in.) glass Pasteur pipette (cotton
plugged, sterile) with the tip slightly flame-constricted—25
up-and-down strokes should be sufficient (see Note 12).
6. Add 3 mL of solution C per tube and apply a further 15 strokes
with the Pasteur pipette.
7. Add 6 mL of solution E per tube.
8. Centrifuge at 200 × g for 7 min.
9. Resuspend the cell pellet in 2–3 mL complete serum-containing
culture medium, using a Pasteur pipette as above. The remaining
steps are the same as steps 10–12 in Subheading 3.1.
3.3. Cell Counting 1. Hemocytometers are modified glass slides engraved with grids
with a Hemocytometer of fixed area at each side of a central trough, which are covered
with a coverslip prior to use. The sample of cells to be counted
is harvested and applied to the slide surface with a Pasteur
pipette or pipetman, and allowed to move under the coverslip
by capillary action.
30 L. Facci and S.D. Skaper
3.4. Setting Up 1. Prepare one or more 96-well tissue culture plastic plates coated
the CGN Cultures with poly-L-lysine.
for Neuroprotection 2. Dilute the CGN cell dissociate to 400,000 cells/mL, using
Assay BME supplemented with 10% heat-inactivated FCS, 2 mM
L-glutamine, 50 μg/mL gentamicin, and 25 mM KCl (see
Note 17).
3. Add to each culture well 0.1 mL of the above cell suspension.
This will give a density of 40,000 CGN/well.
4. Incubate cultures at 37°C in a humidified atmosphere with
5% CO2.
5. Eighteen to twenty-four hours after cell plating, add cytosine
β-D-arabinofuranoside (Ara-C) to a final concentration of
10 μM. To do this, remove 10 μL of medium from each well
and add 10 μL of fresh medium containing 100 μM Ara-C
(diluted 1:100 from a 10-mM stock solution).
3.6. MTT Analysis Ideally, a colorimetric assay for living cells should utilize a colorless
of Cell Viability substrate that is modified to a colored product by any living cell,
of LY294002-Treated but not by dead cells, their lytic debris, or tissue culture medium.
CGN: Protection Tetrazolium salts are attractive candidates for this purpose since
by Corticotropin- they measure the activity of various dehydrogenase enzymes (21).
Releasing Factor The tetrazolium ring is cleaved in active mitochondria, and so the
reaction occurs only in living cells. A rapid colorimetric assay is
based on the tetrazolium salt MTT that measures only living cells
and can be read on a scanning multiwall spectrophotometer (ELISA
reader) (22, 23).
1. At 8 days in culture, exchange the culture medium for serum-
free BME (or Neurobasal-A) containing 25 mM KCl ± 75 μM
LY294002 ± 30 nM corticotropin-releasing factor. After 6, 24,
and 48 h, cellular vitality is evaluated by MTT assay.
2. Remove the culture medium (aspirate under vacuum, using a
Pasteur pipette).
32 L. Facci and S.D. Skaper
3.7. Cell Culture 1. A horizontal laminar flow hood is recommended for dissection
Safety Issues of tissues. Once dissection is complete, all work should be
moved to a Class 2 vertical laminar flow safety cabinet.
2. The antibiotics and enzymes used should be considered as
noxious agents. Suitable precautions should be taken in the use
and disposal of these products.
3. All liquids containing live cells should be aspirated into a waste
vessel containing MicroSol +3 (a broad spectrum disinfectant)
(Anachem), and pipettes placed in containers with MicroSol.
Plasticware is then disposed of via autoclaving.
4. Accumulated culture liquid waste should then be transferred
to a 20-L carboy containing ~100 g of Virkon powder (VK734,
Medisave, UK). Virkon is a multipurpose disinfectant and con-
tains oxone (potassium peroxymonosulfate), sodium dodecyl-
benzenesulfonate, sulfamic acid, and inorganic buffers. Virkon
3 Culture of Rat Cerebellar Granule Neurons and Application… 33
Table 1
Corticotropin-releasing factor protects cerebellar granule
neurons from LY294002-induced death
Treatment MTT
6h
LY 77 ± 6
LY + CRF 92 ± 3∗
24 h
LY 64 ± 5†
LY + CRF 87 ± 6∗∗
48 h
LY 28 ± 1
LY + CRF 84 ± 5∗∗∗
Cerebellar granule neurons were cultured for 8 days, then shifted to serum-free
BME containing 25 mM KCl and 75 μ M LY294002 (“LY”) ± 30 nM
corticotropin-releasing factor (CRF). After the times indicated, cellular vitality
was evaluated by MTT assay. Data are means ± S.D. (n = 3)
∗p < 0.05; ∗∗p < 0.01; ∗∗∗p <0.0001 (LY + CRF vs. LY); p < 0.05 or p < 0.001
vs. LY at 24 and 48 h, respectively; †p < 0.001 vs. LY at 48 h
Adapted (with substantial modification) from Neuropharmacology 45(5), L.
Facci, D.A. Stevens, M. Pangallo, D. Franceschini, S.D. Skaper, P.J.L.M.
Strijbos, Corticotropin-releasing factor (CRF) and related peptides confer
neuroprotection via type 1 CRF receptors, 623–636 (Table 3), Copyright©
(2003), with permission from Elsevier
4. Notes
Acknowledgments
References
1. Raff MC, Barres BA, Burne J, Coles HS, 3-kinase/Akt cell survival pathway. J Biol Chem
Ishizaki Y, and Jacobson, MD (1993) 273, 19929–19932
Programmed cell death and the control of cell 11. D’Mello SR, Galli C, Ciotti C, and Calissano P
survival – Lessons from the nervous system. (1993) Induction of apoptosis in cerebellar
Science 262, 695–700 granule neurons by low potassium: inhibition
2. Hatten ME, and Heintz N (1995) Mechanisms of cell death by insulin-like growth factor I
of neural patterning and specification in the and cAMP. Proc Natl Acad Sci USA 90,
developing cerebellum. Annu Rev Neurosci 10989–10993
18, 385–408 12. Wood KA, Dipasquale B, and Youle RJ (1993)
3. Gallo V, Kingsbury A, Balázs R, and Jørgensen In situ labelling of granule cells for apoptosis-
OS (1987) The role of depolarisation in the associated DNA fragmentation reveals different
survival and differentiation of cerebellar gran- mechanisms of cell loss in developing cerebellum.
ule cells in culture. J Neurosci 7, 2203–2213 Neuron 11, 621–632
4. Lasher RS, and Zagon IS (1972) The effect of 13. Krueger BK, Burne JF, and Raff MC (1995)
potassium on neuronal differentiation in cul- Evidence for large-scale astrocyte death in
tures of dissociated newborn rat cerebellum. the developing cerebellum. J Neurosci 15,
Brain Res 41, 482–488 3366–3374
5. Thangnipon W, Kingsbury A, Webb M, and 14. Bredesen DE (1995) Neuronal apoptosis. Ann
Balázs R (1983) Observations on rat cerebellar Neurol 38, 839–851
granule cells in vitro: influence of substratum, 15. Fernandez-Sanchez MT, and Novelli A (1993)
potassium concentration and relationship Basic fibroblast growth factor protects cerebellar
between neurones and astrocytes. Brain Res neurons in primary culture from NMDA and
313, 177–189 non-NMDA receptor mediated neurotoxicity.
6. Ginham R, Harrison DC, Facci L, Skaper SD, FEBS Lett 335, 124–131
and Philpott K (2001) Upregulation of death 16. Kubo T, Nonomura T, Enokido Y, and
pathways in rat cerebellar granule neurons Hatanaka H (1995) Brain-derived neurotrophic
undergoing apoptosis. Neurosci Lett 302, factor (BDNF) can prevent apoptosis of rat
113–116 cerebellar granule neurons in culture. Dev
7. Miller TM, Tansey MG, Johnson EM Jr, and Brain Res 16, 249–258
Creedon DR (1997) Inhibition of phosphati- 17. Shimoke K, Yamagishi S, Yamada M, Ikeuchi
dylinositol 3-kinase activity blocks depolarisa- T, and Hatanaka H (1999) Inhibition of phos-
tion- and insulin-like growth factor 1-mediated phatidylinositol 3-kinase activity elevates c-Jun
survival of cerebellar granule cells. J Biol Chem N-terminal kinase activity in apoptosis of
272, 9847–9853 cultured cerebellar granule neurons. Dev Brain
8. Cross DAE, Culbert AA, Chalmers KA, Facci Res 112, 245–253
L, Skaper SD, and Reith AD (2001) Selective 18. Crowder RJ, and Freeman RS (2000) Glycogen
small-molecule inhibitors of glycogen synthase synthase kinase-3b activity is critical for neu-
kinase-3 activity protect primary neurones from ronal death caused by inhibiting phosphati-
death. J Neurochem 77, 94–102 dylinositol 3-kinase or Akt but not for death
9. Dudek H, Datta SR, Franke TF, Birnbaum MJ, caused by nerve growth factor withdrawal.
Yao R, Cooper GM, et al (1997) Regulation of J Biol Chem 275, 34266–34271
neuronal survival by the serine-threonine pro- 19. Cross DAE, Alessi DR, Cohen P, Andjelkovich
tein kinase Akt. Science 275, 661–665 M, and Hemmings BA (1995) Inhibition of
10. Pap M, and Cooper GM (1998) Role of glyco- glycogen synthase kinase-3 by insulin mediated
gen synthase kinase-3 in the phosphatidylinositol by protein kinase B. Nature 378, 785–789
3 Culture of Rat Cerebellar Granule Neurons and Application… 37
20. Hetman M, Cavanaugh JE, Kimelman D, and 22. Mosmann T (1983) Rapid colorimetric assay
Xia Z (2000) Role of glycogen synthase kinase- for cellular growth and survival: application to
3β in neuronal apoptosis induced by trophic proliferation and cytotoxicity assays. J Immunol
withdrawal. J Neurosci 20, 2567–2574 Methods 65, 55–63
21. Slater TF, Sawyer B, and Straeuli UD (1963) Studies 23. Manthorpe M, Fagnani R, Skaper SD, and
on succinate-tetrazolium reductase systems. Varon S (1986) An automated colorimetric
III. Points of coupling of four different tetrazo- microassay for neuronotrophic factors. Dev
lium salts. Biochim Biophys Acta 77, 383–393 Brain Res 25, 191–198
Chapter 4
Abstract
The mammalian brain contains undifferentiated, mitotically active, and multipotent neural stem/progenitor
cells that in vivo contribute new neurons and glia to specific areas of the mature brain. When isolated under
the appropriate conditions, these cells maintain in vitro the ability to proliferate and differentiate into cells
that express neuronal and glial markers. Neural stem/progenitor cells have been identified and isolated
from many regions of the embryonic, postnatal, and adult central nervous system, including cerebellum.
This chapter details techniques to isolate and culture neural progenitor cells from rat postnatal cerebellum,
which can be used as an in vitro model to study the molecular mechanisms underlying proliferation and
differentiation into mature neural cells induced by various stimuli including pharmacological agents.
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_4, © Springer Science+Business Media, LLC 2012
39
40 M. Zusso and P. Debetto
2. Materials
2.1. Dissection 1. Animals: Sprague Dawley rat pups; age of pups = postnatal day
and Cell Cultures 7. We define postnatal day 0 as the day of birth.
2. HBSS/BSA: Add 3 mg/mL bovine serum albumin (BSA) to
calcium- and magnesium-free Hank’s balanced salt solution
(HBSS; Gibco, Life Technologies, Rockville, MD) and filter
sterilize through 0.22 mm filter. Store at 4°C for up to
1 month.
3. Trypsin solution: Add 0.8 mg/mL trypsin to HBSS/BSA and
filter sterilize (see Note 1).
4. Trypsin inhibitor solution: Add 0.5 mg/mL trypsin inhibitor
and 0.1 mg/mL deoxyribonuclease I (DNase) to HBSS/BSA.
Filter sterilize (see Note 1).
5. Growth factor stock solutions: Reconstitute epidermal growth
factor (EGF) (human recombinant) and basic fibroblast growth
factor (bFGF) (Sigma-Aldrich) in order to have a 500 mg/mL
4 Isolation and Culture of Neural Progenitor Cells from Rat Postnatal Cerebellum 41
3. Methods
3.1. Dissection 1. Rapidly decapitate 7-day-old rat pups (see Note 5) by cutting
of Cerebellum the neck with sterile scissors large enough to ensure the head is
from Postnatal Rats removed in one cut. Discard the body into a plastic bag for
and Establishment disposal. Transfer the heads to a sterile 100-mm ∅ Petri dish
of Primary Cultures containing cold HBSS/BSA and cut the skin on top of the
head (see Note 6).
2. Under the dissection hood, cut the skull along the midline,
starting from the back of the head to the eyes. Make a superfi-
cial cut and be careful not to damage the underlying brain tis-
sue. Using forceps, lift up the skull to expose the brain.
3. Using a pair of curved forceps, remove the cerebellum and
immediately transfer it to a sterile Petri dish containing cold
HBSS/BSA (see Note 7). Repeat step 2 until all cerebella have
been collected.
4. Under the dissecting microscope, carefully remove the menin-
ges from each cerebellum using two forceps. The meninges
will appear as a delicate membrane containing blood vessels.
Place the cleaned cerebella into a new dish containing cold
HBSS/BSA and keep on ice.
5. Once all the cerebella have been processed, under the tissue
culture hood, remove the HBSS/BSA from the dish and mince
the tissue into small pieces (0.5–1 mm3) with a flamed razor
blade (see Note 8). Transfer dissected tissues into a 50-mL
conical tube containing 1-mL HBSS/BSA/cerebellum and
centrifuge at 1,000 × g for 3 min.
6. Carefully aspirate the HBSS/BSA (see Note 9), resuspend the
tissue pieces in 1-mL trypsin solution/cerebellum, and incu-
bate for 15 min in a 37°C water bath under gentle shaking to
keep the tissue pieces suspended.
7. Stop the enzymatic reaction by adding the trypsin inhibitor
solution (1 mL/cerebellum). Pellet the tissue by centrifuga-
tion at 1,000 × g for 7 min and remove the supernatant by
gentle aspiration (see Note 9).
8. Add 5 mL HBSS/BSA and dissociate the tissue by triturating
with a sterile, fire-polished, cotton-plugged glass Pasteur pipette
4 Isolation and Culture of Neural Progenitor Cells from Rat Postnatal Cerebellum 43
until the suspension looks cloudy and only small pieces of tissue
are left (see Note 10).
9. Pellet the cells by centrifugation at 1,000 × g for 7 min. Discard
the supernatant and resuspend the cells in 10 mL of pre-
warmed (37°C) culture medium. Make an appropriate dilu-
tion of an aliquot of the cell suspension in trypan blue solution
and count the number of viable cells using a hematocytometer
(see Note 11).
10. Plate the cells at a density of 2 × 105 viable cells/cm2 in uncoated
tissue culture plates, in culture medium containing 20 ng/mL
of EGF and bFGF.
11. Incubate at 37°C, 5% CO2, in a humidified incubator.
12. After 5–6 days, replace half of the medium with fresh medium.
Add a double amount of EGF and bFGF to give a final concen-
tration of 20 ng/mL.
Fig. 1. Morphology and characterization of cerebellar neural progenitors. (a) Representative phase contrast photomicrograph
of a cerebellar neural progenitor cell culture maintained in growth medium for 10 days. (b) Fluorescent photomicro-
graph of the specific immunoreactivity for nestin. Cells were replated onto poly-D-lysine-coated chamber slides in differentiation
medium. After 3 days, cells were fixed with 4% paraformaldehyde and stained with specific antibodies. (c, d) Fluorescent
photomicrographs of neurofilament- (NF), glial fibrillary acidic protein- (GFAP), and O4-positive cells. DAPI staining shows
the nuclei. Scale bars = 50 mm (adopted in part from ref. (16), with modifications).
3.4. Immuno- All staining procedures are carried out at room temperature except
cytochemical Staining where indicated. Washing steps are done for 5 min each:
for Characterization
1. Remove culture medium and carefully wash cultures twice
of Differentiation
with PBS.
2. Fix cells for 10 min in 4% paraformaldehyde at room tempera-
ture and wash three times with PBS (see Note 16).
3. Block nonspecific staining by incubation for 1 h in PBS con-
taining 5% normal donkey serum (Jackson ImmunoResearch
Laboratories) and 0.1% Triton X-100 (blocking solution) at
room temperature (see Note 17).
4. Incubate with primary antibodies diluted in blocking solution for
2 h at room temperature or overnight at 4°C (see Note 18).
5. Wash cells three times with PBS and incubate for 1 h at room
temperature in the dark with species-specific secondary anti-
bodies (conjugated to the desired fluorophores) diluted in
blocking solution (see Note 18).
6. Wash cells three times in PBS, incubate in PBS containing
0.1 mg/mL DAPI for 2 min (for nuclear counterstain), and
then coverslip in Fluoromount mounting medium (Sigma-
Aldrich) (see Note 19).
7. Analyze cell phenotype by confocal or fluorescence microscopy.
4. Notes
under a fume hood and wear gloves and mask. Disposal should
follow chemical safety rules.
4. All the antibodies should be freshly diluted just before use. Do
not store them as diluted solutions.
5. For a good cell yield, use at least one large litter of rats (12 or
more). Rats are generally sacrificed by cervical dislocation prior
to decapitation or by decapitation alone (this depends on the
animal care protocols of your institution).
6. In Subheading 3.1, step 1 can be performed at the lab bench.
To prevent contamination, use sterile instruments (autoclaved
using autoclave bags and opened only when needed) and solu-
tions and take precautions to maintain sterility, including the
wearing of gloves. Speed is also important.
7. The Petri dish should be maintained on ice for the duration of
dissection.
8. Chop the tissues with a rapid chopping motion, moving the
dish in a circular fashion so that all tissue pieces are evenly cut.
9. Do not use the vacuum since there is a risk of sucking out the
tissue.
10. Normally, 20–30 strokes are sufficient, but perform a second
trituration of 15 strokes in case undissociated tissue is still vis-
ible. During triturating, always avoid foaming and bubbles.
11. Initially try a 1:5 dilution. The number of viable cells yielded
by this protocol, using a single postnatal day 7 rat cerebellum,
is 10 × 106 cells. Approximately 5–10% of plated cells form pro-
genitor cell aggregates.
12. Always avoid foaming and bubbles.
13. Feed cells with fresh medium (replace half of the medium)
every 5–6 days.
14. Isolated cerebellar neural progenitors can be subcultured for
three passages.
15. Trituration into a single cell suspension is critical in the differ-
entiation protocol. A drop of cell suspension should be added
to a Petri dish to check that the suspension is free from large
aggregates before plating. Trituration in trypsin/EDTA should
be avoided as it damages the cell membrane and can cause
adhesion defects.
16. Fixed cells can be stored in PBS at 4°C for about a week or can
be processed immediately for immunocytochemistry.
17. Make blocking solution with serum from the species in which
the secondary antibodies were raised. Since blocking solution
contains serum, it can be contaminated. Blocking solution is
stable for 1 week at 4°C; if the solution becomes cloudy, it has
become contaminated and should not be used.
4 Isolation and Culture of Neural Progenitor Cells from Rat Postnatal Cerebellum 47
References
1. Gage FH (2000) Mammalian neural stem cells. 10. Altman J, and Bayer SA (1997) Development
Science 287, 1433–1438 of the cerebellar system: in relation to its evolu-
2. Taupin P, and Gage FH (2002) Adult neuro- tion, structure, and functions. CRC Press, Boca
genesis and neural stem cells of the central ner- Raton, Florida, USA
vous system in mammals. J Neurosci Res 69, 11. Fujita S, Shimada M, and Nakamura T (1966)
745–749 H3-Thymidine autoradiographic studies on the
3. Kempermann G, Wiskott L, and Gage FH cell proliferation and differentiation in the exter-
(2004) Functional significance of adult neuro- nal and internal granular layers of the mouse
genesis. Curr Opin Neurobiol 14, 186–191 cerebellum. J Comp Neurol 128, 191–208
4. Morshead CM, Reynolds BA, Craig CG, 12. Altman J (1969) Autoradiographic and histo-
McBurney MW, Staines WA, Morassutti D, logical studies of postnatal neurogenesis. III.
et al (1994) Neural stem cells in the adult Dating the time of production and onset of dif-
mammalian forebrain: a relatively quiescent ferentiation of cerebellar microneurons in rats.
subpopulation of subependymal cells. Neuron J Comp Neurol 137, 433–458
13, 1071–1082 13. Rakic P (1971) Neuron-glia relationship dur-
5. Palmer TD, Takahashi J, and Gage FH (1997) ing cell migration in developing cerebellar
The adult rat hippocampus contains primordial cortex: A Golgi and electron microscopic
neural stem cells. Mol Cell Neurosci 8, 389–404 study in Macacus Rhesus. J Comp Neurol 141,
6. Palmer TD, Markakis EA, Willhoite AR, Safar 283–312
F, and Gage FH (1999) Fibroblast growth fac- 14. Abraham H, Tornoczky T, Kosztolanyi G, and
tor-2 activates a latent neurogenic program in Seress L (2001) Cell formation in the cortical
neural stem cells from diverse regions of the layers of the developing human cerebellum. Int
adult CNS. J Neurosci 19, 8487–8497 J Dev Neurosci 19, 53–62
7. Lie DC, Dziewczapolski G, Willhoite AR, 15. Lee A, Kessler JD, Read TA, Kaiser C, Corbeil
Kaspar BK, Shults CW, and Gage FH (2002) D, Huttner WB, et al (2005) Isolation of neu-
The adult substantia nigra contains progenitor ral stem cells from the postnatal cerebellum.
cells with neurogenic potential. J Neurosci 22, Nat Neurosci 8, 723–729
6639–6649 16. Zusso M, Debetto P, Guidolin D, Barbierato
8. Weiss S, Dunne C, Hewson J, Wohl C, Wheatley M, Manev H, and Giusti P (2008) Fluoxetine-
M, Peterson AC, et al (1996) Multipotent induced proliferation and differentiation of
CNS stem cells are present in the adult mam- neural progenitor cells isolated from rat post-
malian spinal cord and ventricular neuroaxis. J natal cerebellum. Biochem Pharmacol 76,
Neurosci 16, 7599–7609 391–403
9. Klein C, Butt SJ, Machold RP, Johnson JE, and 17. Sottile V, Li M, and Scotting PJ (2006) Stem
Fishell G (2005) Cerebellum- and forebrain- cell marker expression in the Bergmann glia
derived stem cells possess intrinsic regional population of the adult mouse brain. Brain Res
character. Development 132, 4497–4508 1099, 8–17
Chapter 5
Abstract
Neurons cultured from rodent central nervous system tissues represent an important tool in the study of
neurodegenerative disease mechanisms and neuroregenerative processes, including the survival- and axon
growth–promoting properties of neurotrophic factors. This chapter presents a detailed protocol for the
preparation of rat and mouse cortical and hippocampal neuron cell cultures using either embryonic or
postnatal tissue with enzymatic digestion.
Key words: Cortex, Hippocampus, Neurons, Cell culture, Rat, Mouse, Embryonic, Newborn
1. Introduction
The study of central nervous system diseases has been greatly facili-
tated by the use of ex vivo models which are based on the culture
of neurons from discrete brain areas, for example, cortex, hip-
pocampus, and striatum. Procedures have been developed to allow
for the survival and development of cultured central neurons largely
free of unwanted glial cell elements, in particular the now widely
used B27/Neurobasal formulation (1). Such cultures allow one to
investigate in detail the molecular events involved in amyloid
β-peptide toxicity, excitatory amino acid toxicity, oxidative stress,
the survival-promoting effects of growth factors and cell adhesion
molecule mimetics, kinase signaling pathways, and electrophysio-
logical behaviors (2–13). The following chapter describes a proto-
col for the culture of cortical and hippocampal neurons from
embryonic and newborn rodent (rat, mouse) tissues, based on the
B27/Neurobasal serum-free medium.
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_5, © Springer Science+Business Media, LLC 2012
49
50 L. Facci and S.D. Skaper
2. Materials
2.2. Reagents 1. Hank’s balanced salt solution, without CaCl2 and MgCl2,
sterile (HBSS) (Invitrogen)
2. Neurobasal medium (Invitrogen)
3. Neurobasal-A medium (Invitrogen)
4. B27 supplement, 50×, with antioxidants (Invitrogen)
5. B27 supplement, 50×, without antioxidants (Invitrogen)
6. Fetal calf serum (FCS) (Invitrogen) (see Note 1)
7. L-Glutamine (200 mM stock), sterile, for cell culture (Invitrogen)
8. Sodium pyruvate (100 mM stock), sterile, for cell culture (Sigma)
9. Penicillin/streptomycin, 10,000 U/mL penicillin + 10,000 μg/
mL streptomycin (100× stock), sterile, for cell culture
(Invitrogen)
10. MgCl2, 1 M, sterile (Sigma)
11. KCl, 3 M, sterile (Sigma)
12. HEPES, 1 M, sterile (Sigma)
13. Cytosine β-D-arabinofuranoside
14. Papain digestion kit (Worthington Biochemicals)
2.3. Culture Media 1. HBSS for dissection. To 490 mL of calcium- and magnesium-
and Other Solutions free HBSS, add 2.25 g of D-glucose, 5 mL of 100-mM sodium
pyruvate solution, and 5 mL of 1-M HEPES solution. Filter-
sterilize.
5 Culture of Rodent Cortical and Hippocampal Neurons 51
3. Methods
3.1. Dissection of Rat 1. Rat cultures are routinely performed using animals at 18 days
Embryonic Cortex of gestation. Euthanize the pregnant rat (CD strain, Sprague
and Hippocampus Dawley) following appropriate national and/or institutional
guidelines for animal sacrifice (see Note 3).
2. Place the animal dorsal side down and wipe thoroughly with a
tissue soaked in 70% ethanol. Make an incision through the
skin along the midventral line from the sternum to the pubis.
3. Make an incision through the belly wall muscles. While push-
ing away the muscle, pick up one of the uterine horns with
forceps and cut the attachment of the uterine horns to the
dorsal body wall.
4. Place the uterine horns in a 10-cm ∅ Petri dish containing cold
HBSS for dissection.
5. Remove the second uterine horn in a similar manner.
6. Remove the embryos from the uterine segments and place in a
new Petri dish containing ice-cold HBSS for dissection.
7. Decapitate embryos and store heads in HBSS (for dissection)
on ice until/during dissection.
8. Remove brain from head by cutting the scalp along the mid-
brain line with fine scissors.
52 L. Facci and S.D. Skaper
3.2. Papain Digestion 1. The papain dissociation kit contains all needed reagents men-
of Dissected Cortical tioned below except HEPES.
and Hippocampal 2. Add 2 mL of 1-M HEPES buffer to 100 mL of EBSS (vial 1 in
Tissue and kit). This will still be referred to as EBSS.
Preparation of a Single 3. Solution C: Add 32 mL of EBSS to the albumin–ovomucoid
Cell Suspension inhibitor mixture (vial 4 in kit, containing 10 mg ovomucoid
and 10 mg albumin/mL) and allow to dissolve.
4. Solution A: Add 5 mL of EBSS to the papain vial (vial 2 in kit,
containing 20 U of papain/mL in 1 mM L-cysteine with
0.5 mM EDTA).
5. Solution B: Add 500 μL of EBSS to the DNase vial (vial 3 in
kit, giving 2,000 U DNAse/mL) and mix gently.
6. Solution D: To solution A, add 250 μL of solution B.
7. Remove excess HBSS from the cortical tissue and add 5 mL of
solution D.
8. Incubate at 37°C for 15 min with occasional gentle agitation.
9. Vigorously triturate tissue with a long (9-in.) Pasteur pipette
until the cell suspension is devoid of visible clumps.
10. Although not usually necessary, small clumps of tissue remain-
ing can be removed by passing the suspension over a 70-μm
mesh filter (Falcon-BD Biosciences), having first inserted the
strainer into the mouth of a 50-mL conical centrifuge tube.
11. Centrifuge the cell suspension (or filtrate) at 200 × g for 5 min.
5 Culture of Rodent Cortical and Hippocampal Neurons 53
12. While the cells are centrifuging, in a 15-mL tube, mix 2.7 mL
EBSS with 0.3 mL solution C and add 0.15 mL solution B
(“EBSS mix”).
13. Place 5 mL solution C in a second 15-mL conical centrifuge
tube.
14. Following centrifugation, discard the supernatant and resus-
pend the cell pellet in the above “EBSS mix.”
15. Using a 5-mL pipette, carefully layer the cell suspension on top
of the 5 mL ovomucoid solution.
16. Centrifuge the ovomucoid gradient at 150 × g for 5 min.
17. Discard the supernatant and resuspend the cell pellet as required
in Neurobasal medium for embryonic or neonatal cells.
18. Optimal cell attachment and survival can be obtained by pre-
coating the poly-D-lysine culture surface with culture medium
containing 10% (v/v) FCS. It is sufficient to expose the surface
to FCS-containing medium for 1 h at 37°C. The FCS-
containing medium should be removed just prior to cell
plating.
19. Count cells (see Chap. 3) and dilute as required. For a 96-well
plate, recommended density is 35,000 cells/well in 0.1 mL
medium.
20. Average yields are 5–6 × 106 cells and 6–8 × 105 cells/rat embry-
onic day 18 cortex and hippocampus, respectively (see Note 6).
21. In our experience, such cultures can be maintained for up to
1 month (see Note 7).
3.3. Preparation 1. Use newborn pups (both sexes) within 24 h of birth. It is our
of Newborn (Rat, experience that neuron cell survival diminishes markedly if the
Mouse) Cortical tissue is older than 1 day postnatal.
Neuron Cell Cultures 2. Euthanize the pups strictly following appropriate institution-
ally approved guidelines.
3. The dissection differs in some respects from that used for
embryos. Cerebral hemispheres are dissected out following
standard techniques and anatomical landmarks.
4. Following sacrifice and decapitation, place heads on a disinfec-
tant wipe and spray with 70% ethanol.
5. Make an incision under the skin along the midline of the dorsal
surface of the head and peel back the skin.
6. Make a second incision close to the midline along the exposed
dorsal surface (the cut should be along the complete antero-
posterior axis of the lateral ventricles).
7. Splay open the hemisphere along the cut surface and continue
cut along the dorsal surface of the hemisphere.
54 L. Facci and S.D. Skaper
4. Notes
Acknowledgment
References
1. Brewer GJ (1995) Serum-free B27/neurobasal mimetics activate TrkB signaling and prevent
medium supports differentiated growth of neuronal degeneration in rodents. J Clin Invest
neurons from the striatum, substantia nigra, 120, 1774–1785. doi: 10.1172/JCI41356
septum, cerebral cortex, cerebellum, and den- 4. Edwards D, Das M, Molnar P, and Hickman JJ
tate gyrus. J Neurosci Res 42, 674–683 (2010) Addition of glutamate to serum-free
2. Hussain I, Harrison DC, Hawkins J, Chapman culture promotes recovery of electrical activity
T, Marshall I., Facci L, et al (2011) TASTPM in adult hippocampal neurons in vitro. J
mice expressing APP and PS-1 mutant trans- Neurosci Methods 190, 155–163
genes are sensitive to γ-secretase modulation 5. Baptista MS, Melo CV, Armelão M, Herrmann
and Aβ42 lowering by GSM-10h. Neurodege- D, Pimentel DO, Leal G, et al (2010) Role of
nerative Dis 8, 15–24 the proteasome in excitotoxicity-induced cleav-
3. Massa SM, Yang T, Xie Y, Shi J, Bilgen M, age of glutamic acid decarboxylase in cultured
Joyce JN, et al (2010) Small molecule BDNF hippocampal neurons. PLoS One 5, e10139
56 L. Facci and S.D. Skaper
6. Franceschini D, Guisti P and Skaper SD (2006) neurons: a possible role for mast cells. J Neurochem
MEK inhibition exacerbates ischemic calcium 76, 47–55
imbalance and neuronal cell death in rat corti- 10. Chapman GA, Moores K, Harrison D,
cal cultures. Eur J Pharmacol 553, 18–27 Campbell CA, Stewart BR, and Strijbos, PJ
7. Skaper SD, Facci L, Williams G, Williams E-J, (2000) Fractalkine cleavage from neuronal
Walsh FS, and Doherty P (2004) A dimeric membranes represents an acute event in the
version of the short N-cadherin binding motif inflammatory response to excitotoxic brain
HAVDI promotes neuronal cell survival by damage. J Neurosci 20, RC87
activating an N-cadherin/fibroblast growth 11. Skaper SD, Ancona B, Facci L, Franceschini D,
factor receptor signalling cascade. Mol Cell and Giusti P (1998) Melatonin prevents the
Neurosci 26, 17–23 delayed death of hippocampal neurons induced
8. Hughes JP, Staton PC, Wilkinson MG, by enhanced excitatory neurotransmission and
Strijbos PJLM, Skaper SD, Arthur JSC, et al the nitridergic pathway. FASEB J 12, 725–731
(2003) Mitogen and stress response kinase-1 12. Skaper SD, Leon A, and Facci L (1991) Death of
(MSK1) mediates excitotoxic induced death cultured hippocampal pyramidal neurons induced
of hippocampal neurones. J Neurochem 86, by pathological activation of N-methyl-D-
25–32 aspartate receptors is reduced by monosialogan-
9. Skaper SD, Facci L, Kee WJ, and Strijbos PJLM gliosides. J Pharmacol Exp Ther 259, 452–457
(2001) Potentiation by histamine of synaptically- 13. Choi DW (1987) Ionic dependence of gluta-
mediated excitotoxicity in cultured hippocampal mate toxicity. J Neurosci 7, 369–379
Chapter 6
Abstract
Neurons cultured from rodent central nervous system tissues represent an important tool in the study of
neurodegenerative disease mechanisms and neuroregenerative processes, including the survival- and axon
growth-promoting properties of neurotrophic factors. This chapter presents a detailed protocol for the
preparation of rat and mouse cortical and hippocampal neuron cell cultures, using either embryonic or
postnatal tissue.
Key words: Amyloid, Cortical neurons, Neurotoxicity, Lactate dehydrogenase assay, Luminescence
assay, Cell culture, Rat, Embryonic
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_6, © Springer Science+Business Media, LLC 2012
57
58 L. Facci and S.D. Skaper
2. Materials
2.2. Reagents 1. Hank’s balanced salt solution, with CaCl2 and MgCl2, sterile
(HBSS) (Invitrogen).
2. Neurobasal medium (Invitrogen).
3. B27 supplement, 50×, with antioxidants (Invitrogen).
4. B27 supplement, 50×, without antioxidants (Invitrogen).
5. Fetal calf serum (FCS) (Invitrogen) (see Note 1).
6. L-Glutamine (200 mM stock), sterile, for cell culture
(Invitrogen).
7. Sodium pyruvate (100 mM stock), sterile, for cell culture
(Sigma).
8. Penicillin/streptomycin, 10,000 U/mL penicillin + 10,000 μg/
mL streptomycin (100× stock), sterile, for cell culture
(Invitrogen).
6 Amyloid b-Peptide Neurotoxicity Assay Using Cultured Rat Cortical Neurons 59
2.4. Ab42 Solution 1. Weigh the desired amount of Aβ42 into a 3-mL glass weighing
vial (see Notes 2 and 3).
2. In a CL2 cabinet, add the required volume of HFIP to the
Aβ42 (use 400 μL HFIP per mg of Aβ42) (see Note 4).
3. Briefly mix and leave at room temperature for 10 min to allow
peptide to fully dissolve (the solution should be clear at this
stage).
4. Evaporate the HFIP under a gentle stream of air, for example,
by attaching a Pasteur pipette to the tube delivering the air
stream. Perform this operation under a fume cupboard.
5. At this point, a film of peptide should be visible around the
base of the vial.
6. Resuspend the dried Aβ42 peptide in tissue culture grade
DMSO 1 mg peptide in 44 μL DMSO, ensuring that the
DMSO is pipetted around the base of the eppendorf tip where
the peptide film is visible.
7. This produces a solution of 5-mM peptide, to which is now
added 0.1% trifluoroacetic acid (73 μL/mg peptide).
8. The Aβ42 peptide solution is now diluted directly into pre-
warmed (37°C) culture medium, to give a concentration of
80 μM (taking into account the volume contributed by DMSO
and trifluoroacetic acid).
9. The cortical neuron cell cultures receive one-fourth volume of
the Aβ42 solution (80 μM) in medium, to yield a final peptide
concentration of 20 μM in the cultures.
60 L. Facci and S.D. Skaper
3. Methods
A 0 0 0 I I I H H H G G G
B
C
D
E
F
G
H F F F E E E D D D C C C
4. Collect from each well the full amount (100 μL) of culture
medium and set aside.
5. Transfer 50 μL of sample medium to a flat bottom clear micro-
titer clear 96-well plate.
6. Lyse cells by adding to each well 100 μL culture medium
containing 1% lysis solution (provided in the kit). Leave in the
37°C incubator for at least 10 min (see Note 5).
7. Transfer 10 μL of cell lysate to the flat-bottom 96-well plate.
Add 40 μL culture medium to each of these wells.
8. Add 50 μL reconstituted substrate to all wells.
62 L. Facci and S.D. Skaper
0.4
0.3
0.1
0.0
-0.1
0 25 50 75 100 125
LDH (mU/ml)
3.3. ATP Cell Content 1. Allow CellTiter-Glo® substrate and buffer to come to room
Assay Protocol temperature (see Notes 8 and 9).
(CellTiter-Glo® 2. Starting with the 100 mM ATP stock solution, prepare a work-
Luminescent Cell ing stock solution of 1 mM ATP using Neurobasal medium
Viability Assay) with B27 supplement (no antioxidants).
3. Prepare serial dilutions of ATP in the above culture medium,
covering the range 0.003–2 μM. A 96-well template may be
used, analogous to that described above for LDH assay but
modified to accommodate the concentration range of ATP
standard dilutions.
4. Add 100 μL per well of ATP standards (using empty wells but
in the same 96-well plate with cells).
5. Add to each well (cells or ATP standards) 100 μL of CellTiter-
Glo solution.
6. Allow at least 10 min for cell to lyse and come to equilibrium.
7. Read plate in a Wallac 1450 MICROBETA PLUS liquid
scintillation counter (TopCount) or similar instrument for
luminescence.
8. Calculate ATP values from the standard curve (see Fig. 2 for a
“representative” example).
6 Amyloid b-Peptide Neurotoxicity Assay Using Cultured Rat Cortical Neurons 63
200000
Luminescence
100000
-100000
0 500 1000 1500 2000 2500
ATP (nM)
4. Notes
Acknowledgments
References
1. Glenner GG, and Wong CW (1984) Alzheimer’s 3. Selkoe DJ (1994) Normal and abnormal
disease: initial report of the purification and biology of the beta-amyloid precursor protein.
characterization of a novel cerebrovascular Annu Rev Neurosci 17, 489–517
amyloid protein. Biochem Biophys Res Comm 4. Jarrett JT, Berger EP, and Lansbury PT Jr (1993)
120, 885–890 The carboxy terminus of the beta amyloid protein
2. Goedert M, Wischik CM, Crowther RA, Walker is critical for the seeding of amyloid formation:
JE, and Klug A (1988) Cloning and sequenc- implications for the pathogenesis of Alzheimer’s
ing of the cDNA encoding a core protein of disease. Biochemistry 32, 4693–4697
the paired helical filament of Alzheimer 5. McGowan E, Pickford F, Kim J, Onstead L,
disease: identification as the microtubule- Eriksen J, Yu C, et al (2005) Aβ42 is essential
associated protein tau. Proc Natl Acad Sci USA for parenchymal and vascular amyloid deposi-
85, 4051–4055 tion in mice. Neuron 47, 191–199
6 Amyloid b-Peptide Neurotoxicity Assay Using Cultured Rat Cortical Neurons 65
6. Dahlgren KN, Manelli AM, Stine Jr WB, Baker non-peptide p75NTR ligands inhibit Aβ-induced
LK, Krafft GA, and LaDu MJ (2002) neurodegeneration and synaptic impairment.
Oligomeric and fibrillar species of amyloid-β- PLoS One 3, e3604
peptides differentially affect neuronal viability. 12. Liu F, Gong X, Zhang G, Marquis K, Reinhart
J Biol Chem 277, 32046–32053 P, and Andree TH (2005) The inhibition of
7. Walsh DM, Klyubin I, Fadeeva JV, Cullen WK, glycogen synthase kinase 3β by a metabotropic
Anwyl R, Wolfe MS, et al (2002) Naturally glutamate receptor 5 mediated pathway confers
secreted oligomers of amyloid β protein neuroprotection to Aβ peptides. J Neurochem
potently inhibit hippocampal long-term poten- 95, 1363–1372
tiation in vivo. Nature 416, 535–539 13. Chen J, Zhou Y, Mueller-Steiner S, Chen LF,
8. Scheuner D, Eckman C, Jensen M, Song X, Kwon H, Yi S, Mucke L, et al (2005) SIRT1
Citron M, Suzuki N, et al (1996) Secreted protects against microglia-dependent amyloid-
amyloid beta-protein similar to that in the β toxicity through inhibiting NF-κB signaling.
senile plaques of Alzheimer’s disease is increased J Biol Chem 280, 40364–40374
in vivo by the presenilin 1 and 2 and APP muta- 14. Massa SM, Yang T, Xie Y, Shi J, Bilgen M,
tions linked to familial Alzheimer’s disease. Nat Joyce JN, et al (2010) Small molecule BDNF
Med 2, 864–870 mimetics activate TrkB signaling and prevent
9. Bozyczko-Coyne D, O’Kane TM, Wu ZL, neuronal degeneration in rodents. J Clin
Dobrzanski P, Murthy S, Vaught JL, et al Invest 120, 1774-1785. doi: 10.1172/
(2001) CEP-1347/KT-7515, an inhibitor of JCI41356
SAPK/JNK pathway activation, promotes sur- 15. Pym LJ, Buckingham SD, Tsetlin V, Boyd CA,
vival and blocks multiple events associated with and Sattelle DB (2007) The Aβ1-42M35C
Abeta-induced cortical neuron apoptosis. J mutated amyloid peptide Aβ1-42 and the 25-35
Neurochem 3, 849–863 fragment fail to mimic the subtype-specificity
10. Hussain I, Harrison DC, Hawkins J, Chapman of actions on recombinant human nicotinic
T, Marshall I, Facci L, et al (2011) TASTPM acetylcholine receptors (α7, α4β2, α3β4).
mice expressing APP and PS-1 mutant trans- Neurosci Lett 427, 28–33
genes are sensitive to γ-secretase modulation 16. Wogulis M, Wright S, Cunningham D, Chilcote
and Aβ42 lowering by GSM-10 h. Neurodegener T, Powell K, and Rydel RE (2005) Nucleation-
Dis 8, 15–24 dependent polymerization is an essential com-
11. Yang T, Knowles JK, Lu Q, Zhang H, Arancio ponent of amyloid-mediated neuronal cell
O, Moore LA, et al (2008) Small molecule, death. J Neurosci 25, 1071–1080
Chapter 7
Abstract
The protocol described in this chapter covers the preparation and culture of enriched populations of
microglia, astrocytes, and oligodendrocytes from the cortex and spinal cord of neonatal rat and mouse.
The procedure is based on the enzymatic digestion of tissue, followed by the culture of a mixed glial cell
population which is then utilized as the starting point for the isolation, via differential attachment, of the
different cell types.
Key words: Microglia, Astrocytes, Oligodendrocytes, Cortex, Spinal cord, Rat, Cell culture
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_7, © Springer Science+Business Media, LLC 2012
67
68 S.D. Skaper et al.
2. Materials
3. Methods
3.1. Dissection of Rat 1. Rat cultures are routinely performed using animals at postnatal
Cortical Tissue day 1–2. Following appropriate national and institutional
guidelines for animal sacrifice, pregnant rats (CD strain,
Sprague Dawley) are euthanized (see Note 1).
2. Dissect out cerebral hemispheres following standard tech-
niques and anatomical landmarks. The procedure is applicable
to either rat or mouse pups.
3. Make an incision under the skin along the midline of the dorsal
surface and peel the skin back.
4. Make a second incision close to the midline along the exposed
dorsal surface (the cut should be along the complete antero-
posterior axis of the lateral ventricles).
5. Splay open the hemisphere along the cut surface and continue
the cut along the dorsal surface of the hemisphere.
7 Culture of Neonatal Rodent Microglia, Astrocytes, and Oligodendrocytes… 71
3.2. Enzymatic 1. Grip a straight-edge razor blade with a hemostat, dip in 70%
Digestion of Cortical ethanol, and pass through a flame to sterilize.
Tissue and 2. Mince the collected cortical tissue with the razor blade.
Preparation of Cell
3. Collect the minced tissue with L-15 and transfer to a 15-mL
Suspension for Plating centrifuge tube.
4. Centrifuge collected cortical tissue for 30 s (200 × g) to com-
pact the minced tissue.
5. While the tissue is centrifuging, prepare the following: 3 mL of
L-15 medium containing 140 μL stock papain solution (as pur-
chased), 30 μL of 100× stock L-cysteine solution, and 30 μL of
100× DNase stock solution. Filter-sterilize before use.
6. Remove L-15 medium from the tube with cortical tissue pel-
let, and add the L-15 solution containing papain, DNase, and
L-cysteine.
15. Using a 5-mL tissue culture pipette, resuspend the cell pellet in
3 mL plating medium. Add an additional amount of plating
medium such that the final volume is equivalent to 2 mL × num-
ber of T-75 flasks to be used (e.g., 20 mL final for 10 flasks).
16. Add 18 mL plating medium to each flask, followed by 2 mL of
cell suspension. Normally, we seed cultures at a ratio of 1.5
brains/flask (see Note 5).
17. The following day, replace the culture medium with 15 mL
fresh plating (growth) medium.
18. Twice per week, replace one-half the volume of culture medium
with an equal volume of fresh growth medium.
19. The mixed glial cell cultures are incubated for 10–14 days,
after which time, confluence will have been reached and they
can be taken for harvesting different cell populations (astro-
cytes, microglia, oligodendrocytes) (see Note 6).
3.3. Isolation 1. Remove 3 mL medium from each T-75 flask; this should leave
of Oligodendrocyte about 12 mL of medium.
Precursors and 2. As the culture medium is bicarbonate-buffered, to prevent pH
Microglia excursions, close the flask cap (or cover with parafilm if the cap
is of the filter type).
3. Place the flasks on an orbital shaker fitted with a temperature-
controlled chamber. Flasks can be fastened to the shaker plat-
form using double-sided adhesive tape, or taped directly to the
platform using high-strength industrial ducting tape.
4. Shake the flasks for 2 h at 200 cycles/min (37°C).
5. Remove the culture medium (containing mainly microglia)
from the flasks and transfer to 10-cm ∅ Sterilin plastic petri
dishes. A volume of 20–25 mL/dish is best (see Notes 7
and 8).
6. Add 13 mL maintenance medium to the flasks and place on
the orbital shaker overnight at 200 cycles/min.
7. Place the Sterilin dishes in the 5% CO2 incubator (37°C) and
leave for 45–60 min.
8. Aspirate the medium on the Sterilin plates and add 6 mL fresh
maintenance medium.
9. Use a cell scraper to remove the attached microglia from the
Sterilin dishes. Transfer the medium with cells to a 50-mL cen-
trifuge tube. Rinse the dishes with 5 mL maintenance medium
and transfer to the collection tube.
10. Resuspend the microglia cell pellet in maintenance medium and
plate into poly-L-lysine-coated multiwall plates as dictated by the
experimental design. Typically, we plate 105 cells in a 96-well
plate (equivalent to 3.5 × 105 cells/cm2). For cell counting
7 Culture of Neonatal Rodent Microglia, Astrocytes, and Oligodendrocytes… 73
3.4. Isolation of an 1. The T-75 flasks remaining after shaking to recover oligoden-
Enriched Population drocytes and microglia are used as a source of highly enriched
of Astrocytes astrocytes.
2. Aspirate the medium from the flask and rinse the cell mono-
layer with 10-mL sterile phosphate-buffered saline.
3. Add to the flask 3 mL of 0.25% trypsin/EDTA solution, rock
the flask to spread the trypsin solution over the entire mono-
layer, and then remove by aspiration. This will leave a thin film
of trypsin solution over the cells.
4. Incubate the flask(s) at 37°C for 10 min.
5. Tap the flask against the palm of the hand to dislodge cells and
add 5 mL maintenance medium per flask; the fetal calf serum
in the medium inactivates the trypsin.
6. Rinse the flask with the medium added, collect cells, and trans-
fer to a 15- or 50-mL centrifuge tube, as needed.
7. Pellet cells by centrifugation at 200 × g for 5 min, re-suspend
pellet in maintenance medium and count. The typical cell yield
is 4–5 × 106 cells/T-75 flask. On this basis, dilute the cell sus-
pension to 1–1.5 × 106 cells/mL for ease of counting. Astrocytes
thus obtained are ³95% pure.
74 S.D. Skaper et al.
3.5. Preparation 1. The procedure is essentially as that described for cortical tissue,
of Spinal Cord Glia except for the dissection.
2. Sacrifice pups by decapitation.
3. Place the body face down on a paper towel. Spray 70% ethanol
over the back.
4. Remove the back skin with surgical scissors to expose the ver-
tebral column.
5. Entering from the anterior side of the body, make a cut through
the midline as far as the tail.
6. Using a fine pair of surgical scissors trim back, at a 45° angle,
the tissue on either side of the spinal canal (being careful not
to disrupt the spinal cord itself).
7. Carefully strip off the spinal cord with a fine pair of forceps and
transfer to a 10-cm ∅ dish of cold L-15 medium.
8. Repeat this procedure for the remaining pups.
9. Peel off the meninges using fine tipped forceps, and transfer
the tissue pieces to a 15-mL centrifuge tube. It is not necessary
to mince the tissue as for cortex; the spinal cord is much smaller
and will already have been fragmented to some extent in the
course of removing the meninges.
10. Centrifuge the collected tissue for 30 s (200 × g) to compact.
11. Remove L-15 medium from the tube and add the L-15 solu-
tion containing papain, DNase, and L-cysteine.
12. Incubate tissue for 60 min in a 37°C water bath with occa-
sional swirling.
13. Proceed with tissue dissociation as for cortex, except the final
5-min centrifugation (Subheading 3.2, step 14). Doing so
reduces the risk of losing material, which is limited in compari-
son to cortex.
14. Following dissociation, plate the spinal cord cell suspension in
medium and distribute in poly-L-lysine-coated T-75 flasks, five
spinal cords per flask.
15. For harvesting microglia, we find that spinal cord mixed glia
cultures mature more slowly than do the cortical cultures. As
such, optimal recovery of microglia is achieved after 10–11
days in vitro. Isolation of separate populations of microglia,
astrocytes, and oligodendrocytes is carried out as described for
cortical cultures.
7 Culture of Neonatal Rodent Microglia, Astrocytes, and Oligodendrocytes… 75
4. Notes
References
1. Schwab ME, and Schnell L (1991) Channeling 9. Culbert AA, Skaper SD, Howlett DR, Evans
of developing rat corticospinal tract axons by NA, Facci L, Soden PE, et al (2006) MAPKAP
myelin-associated neurite growth inhibitors. kinase 2 deficiency in microglia inhibits pro-
J Neurosci 11, 709–721 inflammatory mediator release and resultant
2. Rosin C, Colombo S, Calver AA, Bates TE, neurotoxicity: Relevance to neuroinflammation
and Skaper SD (2005) Dopamine D2 and D3 in a transgenic mouse model of Alzheimer’s
receptor agonists limit oligodendrocyte injury disease. J Biol Chem 281, 23658–23667
caused by glutamate oxidative stress and oxygen/ 10. Skaper SD, Facci L, Culbert A, Evans NA,
glucose deprivation. Glia 52, 336–343 Chessell I, Davis JB, et al (2006) P2X7 recep-
3. Rosin C, Bates TE, and Skaper SD (2004) tors on microglial cells mediate injury to corti-
Excitatory amino acid induced oligodendro- cal neurons in vitro. Glia 54, 234–242
cyte cell death in vitro: receptor-dependent 11. Varon S, Skaper SD, Barbin G, Selak I, and
and-independent mechanisms. J Neurochem Manthorpe M (1984) Low molecular weight
90, 1173–1185 agents support survival of cultured neurons
4. Matute C, Alberdi E, Domercq M, Perez-Cerda from the central nervous system. J Neurosci 4,
F, Perez-Samartin A, and Sanchez-Gomez MV 654–658
(2001) The link between excitotoxic oligoden- 12. Shen LH, Li Y, and Chopp M (2010) Astrocytic
droglial death and demyelinating diseases. endogenous glial cell derived neurotrophic fac-
Trends Neurosci 24, 224–230 tor production is enhanced by bone marrow
5. Gallo V, and Ghiani CA (2000) Glutamate stromal cell transplantation in the ischemic
receptors in glia: new cells, new inputs and boundary zone after stroke in adult rats. Glia
new functions. Trends Pharmacol Sci 21, 58, 1074–1081
252–258 13. Skaper SD, Facci L, and Leon A (1995)
6. Skaper SD, Evans NA, Soden PE, Rosin C, Inflammatory mediator stimulation of astro-
Facci L, and Richardson JC (2009) Oligoden- cytes and meningeal fibroblasts induces neu-
drocytes are a novel source of amyloid peptide ronal degeneration via the nitridergic pathway.
generation. Neurochem Res 34, 2243–2250 J Neurochem 64, 266–276
(doi: 10.1007/s11064-009-0022-9) 14. Miller G (2005) The dark side of glia. Science
7. Trang T, Beggs S, Wan X, and Salter MW 308, 778–781
(2009) P2X4-receptor-mediated synthesis and 15. Facci L, Skaper SD, Favaron M, and Leon A
release of brain-derived neurotrophic factor (1988) A role for gangliosides in astroglial cell
in microglia is dependent on calcium and differentiation in vitro. J Cell Biol 106, 821–828
p38-mitogen-activated protein kinase activation. 16. Gabellini N, Facci L, Milani D, Negro A,
J Neurosci 29, 3518–3528 Callegaro L, Skaper S.D, et al (1991)
8. Zhang P, Hatter A, and Liu B (2007) Differences in induction of c-fos transcription
Manganese chloride stimulates rat microglia to by cholera toxin-derived cyclic AMP and Ca2+
release hydrogen peroxide. Toxicol Lett 173, signals in astrocytes and 3 T3 fibroblasts. Exp
88–100 Cell Res 194, 210–217
7 Culture of Neonatal Rodent Microglia, Astrocytes, and Oligodendrocytes… 77
17. Hamby ME, Uliasz TF, Hewett SJ, and Hewett to the lysosomotropic agent, L-leucine methyl
JA (2006) Characterization of an improved ester. J Immunol 131, 2282–2290
procedure for the removal of microglia from 19. Giulian D, Vaca K, and Corpuz M (1993)
confluent monolayers of primary astrocytes. Brain glia release factors with opposing actions
J Neurosci Methods 150, 128–137 upon neuronal survival. J Neurosci 13, 29–37
18. Thiele DL, Kurosaka M, and Lipsky PE (1983) 20. Hewett JA, Hewett SJ, Winkler S, and Pfeiffer SE
Phenotype of the accessory cell necessary for (1999) Inducible nitric oxide synthase expression
mitogen-activated T and B cell responses in human in cultures enriched for mature oligodendrocytes
peripheral blood: delineation by its sensitivity is due to microglia. J Neurosci Res 56, 189–198
Chapter 8
Abstract
Glial cell activation plays an important role in the pathogenesis of various neurodegenerative disorders.
This article presents a protocol for the preparation of cultures consisting of rat embryonic cortical neurons
grown in the presence of cortical microglia, in which the glia are present in physical contact with the neu-
rons or separated by a semi-permeable membrane barrier.
Key words: Neurons, Microglia, Astrocytes, Cortex, Co-culture, Rat, Cell culture
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_8, © Springer Science+Business Media, LLC 2012
79
80 S.D. Skaper and L. Facci
2. Materials
2.2. Reagents 1. Hank’s balanced salt solution (HBSS), with CaCl2 and MgCl2
(Invitrogen).
2. Neurobasal medium (Invitrogen).
8 Central Nervous System Neuron-Glia Co-culture Models 81
2.3. Culture Media 1. Culture medium for neurons : Neurobasal medium containing
and Solutions B27 supplement (1×), 2 mM L-glutamine, 100 U/mL penicillin,
and 100 μg/mL streptomycin: To 500 mL Neurobasal
medium, add 10 mL 50× B27 supplement, 5 mL 200 mM
glutamine, and 5 mL 10,000 U/ml penicillin to 10,000 μg/mL
streptomycin. Complete medium can be stored at +4°C for up
to 2 months.
2. Culture medium for microglia: To a 500-mL bottle of DMEM,
add 50 mL FCS (10% final), 5 mL stock penicillin/streptomy-
cin (100 U/mL penicillin + 100 μg/mL streptomycin final),
and 0.5 mL of gentamicin stock (50 μg/ml final).
3. 0.15 M borate buffer pH 8.4: Dissolve 28.6 g of sodium borate
(Na2B4O7·10H2O) in 500 mL water (pH will be ~9.2). Adjust
pH to 8.4 with 5 N HCl. Filter-sterilize and store at 4°C (up
to 6 months).
3. Methods
3.1. Neuron-Glia 1. Prepare primary cerebral cortical neuron cell cultures from
Co-cultures: Physical gestational day 18 fetuses of time-mated CD rats (Charles
Contact Model River) (see Chap. 5).
2. Re-suspend the final cell pellet in Neurobasal medium contain-
ing 2% B27 supplements, 1 mM sodium pyruvate, 2 mM
L-glutamine, and penicillin (100 units/mL)/streptomycin
(100 μg/mL).
3. Plate cells in poly-D-lysine-coated 48-well plates, 1 × 105 cells/
well in 0.35 mL of the same medium. Place cultures in a
humidified incubator at 37°C in a 5% CO2/95% air
atmosphere.
4. Expose the poly-D-lysine-coated culture wells overnight to
medium containing 10% FCS prior to cell seeding.
5. After 2 days, add to each culture well 0.35 mL of plating
medium, but containing B27 supplement without antioxidants.
Neuron cell cultures cells are normally used on day 6 for
experiments.
6. Culture microglia from the cortex of newborn CD (Charles
River) rat pups, as described in Chap. 7.
7. Collect microglia from the 10-cm ∅ Sterilin plates by mechan-
ically scraping into culture medium for glia (see Chap. 7).
8. Pellet microglia by centrifugation (200 × g for 5 min) and
re-suspend the cell pellet in Neurobasal medium containing
2% B27 supplements (without antioxidants), 1 mM sodium
pyruvate, 2 mM L-glutamine, and penicillin/streptomycin.
Use 1 mL medium for each Sterilin plate of microglia collected,
and re-suspend pellet using a long (9-in.) Pasteur pipette
attached to an automatic pipettor. Eight or nine up-and-down
strokes are usually sufficient to fully disperse the cell pellet.
Determine the cell density in the suspension by counting with a
hemocytometer (Sec Chap. 3).
9. To prepare the co-cultures, dilute the microglia cell suspension
to 285,000 cells/mL, using the same medium for suspension of
the starting cell pellet.
10. Take the 48-well plate of neurons set up 6 days ago and remove
0.35 mL of medium. Now add to each well 0.35 mL of the
8 Central Nervous System Neuron-Glia Co-culture Models 83
3.2. Assessment 1. LDH release into the culture medium was used as a measure of
of Neurotoxicity cell death and has been utilized previously to quantify neuronal
in Physical Contact cell injury in microglia-neuron co-cultures (8, 9).
Neuron-Microglia 2. LDH activity in the cell culture supernatants is determined
Co-cultures after 72 h, using the CytoTox® non-radioactive cytotoxicity
assay kit.
3. Collect 100 μL of medium from each well of the co-cultures
for assay. Use 50 μL of this sample for assay, and proceed with
the assay as described in Chap. 6 (see Note 4).
4. Express values of LDH release as milliunits/mL (see Notes 5
and 6).
5. Establish separate cultures of microglia without neurons in
poly-D-lysine-coated 48-well plates (100,000 cells/well) in
0.7 mL Neurobasal/B27 (antioxidant-free) at the time micro-
glia are added to the neuron cultures. Incubate these microglia
cultures for 72 h in the presence of stimuli, and then measure
LDH release from the microglia alone ± stimuli.
6. Subtract the microglia only LDH values obtained from the LDH
values obtained for the neuron-microglia co-cultures receiving
the same treatments (see Note 7). The purpose of this step is to
correct for the contribution of microglia LDH release.
3.3. Neuron-Glia 1. This model allows one to culture glia on a porous membrane,
Co-cultures: Transwell which is then placed in a well containing an established mono-
Insert Model layer of neurons. Application of a stimulus to the glia layer
allows for glia-derived agents to diffuse across the membrane
and act on the underlying neurons. The insert with glia can
then be removed, permitting morphological or biochemical
analysis of the neurons. Figure 1 shows a typical commercially
84 S.D. Skaper and L. Facci
Transwell Insert
Microglial Cells
Neurons
15. Transfer the inserts to the 24-well plate of cortical neurons (set
up 6 days ago), without changing the medium in the well.
The porous membrane allows free diffusion of molecules.
The distance between the neuron monolayer and microglia
on the insert membrane is 1 mm. Return the plate to the 5%
CO2 incubator.
16. The insert co-cultures are now ready for use. The treatment
regimen will, of course, be determined by the experimental
plan one wishes to carry out. For example, one can treat the
upper (microglia-containing) chamber with amyloid β-peptide,
using a concentration sufficient to stimulate microglia without
causing injury to the underlying neurons (10), in the presence
86 S.D. Skaper and L. Facci
3.4. Assessment 1. At the end of the incubation period, remove the microglia-
of Neurotoxicity containing inserts and evaluate neuronal cell viability by the
in Transwell Insert colorimetric method based upon conversion of 3-(4,5-dimeth-
Co-cultures ylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) by
mitochondrial dehydrogenases to a blue formazan product.
2. Add to each well 400 μL of phenol red-free Neurobasal
medium containing 2% B27 supplements (without antioxidants),
1 mM sodium pyruvate, 2 mM L-glutamine, and penicillin/
streptomycin, plus 0.15 mg/mL MTT (which represents a 1:20
dilution of the 3 mg/mL stock). We find that this MTT concen-
tration gives optimal color yield (under these conditions).
3. Incubate the MTT-containing cultures for 1 h at 37°C. This
incubation time gives optimal color yield (under these
conditions).
4. Examination of the cultures under a light microscope will show
that viable cells contain blue crystals. This is the formazan
reaction product.
5. Remove the culture medium by aspiration, and add 400 μL of
dimethyl sulfoxide per well. This is to dissolve the reaction
product. Gently rock the plate to mix the blue reaction product
uniformly with the solvent. This should take only 1–2 min.
6. Transfer 100 μL of the colored dimethyl sulfoxide solution
from each well to the well of a clear polystyrene plastic 96-well
microplate (any brand will do). Read the plate on a microplate
(ELISA) reader. We use a SpectraMax M2 microplate reader
from Molecular Devices, although there are a number of qual-
ity plate readers on the market. For optimal results, the plate
should be read at a wavelength of 570 nm (test wavelength),
followed by 630 nm (reference wavelength). The difference
(A570−A630) is used as the final absorbance value generated by
the sample. Correcting for optical imperfections in the
microplates by subtracting A630 is recommended but is not an
essential procedure.
7. Normalize the absolute MTT values by scaling to the mean of
cultures which have not been treated with test compound
(defined as 100%).
8. Cell vitality should be routinely confirmed by morphological
observation using a light microscope with phase contrast optics
(see Note 9).
8 Central Nervous System Neuron-Glia Co-culture Models 87
4. Notes
Acknowledgments
References
1. Griffin WS, Sheng JG, Royston MC, Royston 4. Skaper SD (2010) Microglia as a target for
MC, Gentleman SM, McKenzie JE, et al (1998) inflammatory processes and neuroprotective
Glial-neuronal interactions in Alzheimer’s disease: strategies. Am J Neuroprotec Neuroregen 2,
the potential role of a “cytokine cycle” in dis- 35–47
ease progression. Brain Pathol 8, 65–72 5. Skaper SD (2011) Ion channels on microglia:
2. Colton CA, and Wilcock DM (2010) Assessing therapeutic targets for neuroprotection. CNS
activation states in microglia. CNS Neurol Neurol Disord Drug Targets, 10, 44–56
Disord Drug Targets 9, 174–191 6. Chao CC, Hu S, Molitor TW, Shaskan EG, and
3. Craft JM, Watterson DM, and Van Eldik LJ Peterson PK (1992) Activated microglia medi-
(2005) Neuroinflammation: a potential therapeu- ate neuronal cell injury via a nitric oxide mech-
tic target. Expert Opin. Ther Targets 9, 887–900 anism. J Immunol 149, 2736–2741
8 Central Nervous System Neuron-Glia Co-culture Models 89
7. Flavin MP, Zhao G, and Ho LT (2000) 10. Culbert AA, Skaper SD, Howlett DR, Evans
Microglial tissue plasminogen activator (tPA) NA, Facci L, Soden PE, et al. (2006) MAPKAP
triggers neuronal apoptosis in vitro. Glia 29, kinase 2 deficiency in microglia inhibits pro-
347–354 inflammatory mediator release and resultant
8. Golde S, Chandran S, Brown GC, and neurotoxicity: Relevance to neuroinflammation
Compston A (2002) Different pathways for in a transgenic mouse model of Alzheimer’s
iNOS-mediated toxicity in vitro dependent on disease. J Biol Chem 281, 23658–23667
neuronal maturation and NMDA receptor 11. Skaper SD, Facci L, Culbert A, Evans NA,
expression. J Neurochem 82, 269–282 Chessell I, Davis JB, et al. (2006) P2X7 recep-
9. Parvathenani LK, Tertyshnikova S, Greco tors on microglial cells mediate injury to corti-
CR, Roberts SB, Robertson B, and Pos- cal neurons in vitro. Glia 54, 234–242
mantur R (2003) P2X 7 mediates superoxide 12. Jana A, and Pahan K (2010) Fibrillar amyloid-
production in primary microglia and is up- β-activated human astroglia kill primary human
regulated in a transgenic mouse model neurons via neutral sphingomyelinase: implica-
of Alzheimer’s disease. J Biol Chem 278, tions for Alzheimer’s disease. J Neurosci 30,
13309–13317 12676–12689
Chapter 9
Abstract
Dopaminergic neuronal cell degeneration is the principal characteristic feature of the neuropathology of
Parkinson’s disease. Cultures of mesencephalic neurons are widely used as a source of dopaminergic neu-
rons for the study of mechanisms implicated in dopaminergic cell death and for the evaluation of potential
dopaminergic neuroprotective agents, including neurotrophic factors. This chapter presents a detailed
protocol for the preparation of rat mesencephalic cell cultures and their application to evaluating the neu-
roprotective action of brain-derived neurotrophic factor.
Key words: Mesencephalic, Ventral tegmentum, Rat, Brain-derived neurotrophic factor, Dopamine
uptake, Tyrosine hydroxylase, Parkinson’s disease
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_9, © Springer Science+Business Media, LLC 2012
91
92 S.D. Skaper et al.
2. Materials
2.2. Culture Media, 1. Hank’s balanced salt solution (HBSS, with CaCl2 and MgCl2)
Supplements, (Invitrogen).
and Additives 2. Dulbecco’s modified Eagle’s medium, high glucose with
GlutaMAX I, and sodium pyruvate (Invitrogen).
3. Neurobasal medium (Invitrogen).
4. B27 supplement (with antioxidants, 50×) (Invitrogen).
5. B27 supplement (without antioxidants, 50×) (Invitrogen).
6. N2 supplement (100×) (Invitrogen).
7. Fetal calf serum (Invitrogen).
8. L-Glutamine (200 mM, sterile, for cell culture) (Invitrogen).
9. Non-essential amino acids (100×, sterile) (Invitrogen).
10. Penicillin/streptomycin (100×, sterile) (Invitrogen).
11. Poly-D-lysine (MW 30,000–70,000, sterile, for cell culture)
(Sigma).
12. [3H]Dopamine (DA) (Amersham Biosciences).
13. [γ-14C]Aminobutyric (GABA) (Amersham Biosciences).
9 Culture and Characterization of Rat Mesencephalic Dopaminergic Neurons 93
3. Methods
3.1. Isolation 1. Euthanize pregnant rats (CD strain, Sprague Dawley) following
of Mesencephalic appropriate national and institutional guidelines. Under asep-
Cells tic conditions, remove the whole brain from the rat embryos
(gestational age 14–15 days) and collect in a petri dish (10 cm
∅) containing ice-cold Medium 4. Remove the blood vessels
and meninges.
2. After having collected all brains, dissect out the ventral two-
thirds of the mesencephalon. The dissected regions include
dopaminergic neurons in the substantia nigra and the ventral
tegmental area but not the noradrenergic neurons in the locus
ceruleus.
3. When all cortices have been dissected, mechanically dissociate
the tissue in 2 mL of Neurobasal medium containing 10%
(v/v) fetal calf serum, using a series of three flame-polished
Pasteur pipettes of decreasing bore size (see Notes 4 and 5).
4. Count the cell suspension (see Chapter 3). A typical yield is
106 viable cells per embryo.
5. Use a poly-L-lysine substratum for culture. The method of
preparation follows that described in Chapter 3 (see Note 6).
6. Centrifuge the cell suspension (200 × g, 5 min), remove the
supernatant, and resuspend the cell pellet in prewarmed (37°C)
Medium 1. Plate at 1,100–2,500 cells/mm2.
7. After 5 days in culture, replace 1/2 of the plating medium with
Medium 2 (this may not be necessary—check pH).
8. Replace 1/2 of the culture medium twice a week with Medium
2, until the cultures are used.
3.4. High-Affinity [14C] 1. Incubate mesencephalic cells at 37°C for 15 min with 0.1 μM
GABA Uptake [14C]GABA in Krebs-Ringer buffer in the presence of 2 mM
β-alanine (1:250 from a 0.5 M stock solution in H2O), an
inhibitor of glial high-affinity GABA uptake.
2. Verify high-affinity neuron-specific (14C)GABA uptake using
the inhibitor L-2,4-diaminobutyric acid (1 mM final, 1:500
from a 0.5 M stock in H2O).
3. Washing and extraction procedures are as for (3H)DA uptake.
6000 **
4000
3000
*
2000
1000
0
Ctrl BDNF TOPA TOPA+ TOPA+ TOPA+ TOPA+
BDNF BDNF BDNF BDNF
(1) (3) (10) (50)
Fig. 2. BDNF protects cultured mesencephalic dopaminergic neurons from the neurotoxic
effects of TOPA. After 24 h in vitro, cultures received BDNF (1–50 ng/mL). Cultures were
kept until seven DIV, before being exposed to 10 μM TOPA for 24 h. At the end of the
experiment (8 days in total), all cover glasses were processed for TH immunoreactivity,
and the number of TH+ cells was determined in each group (mean ± SD, n = 6, 3 experi-
ments). Values in parentheses indicate BDNF concentration (ng/mL). *P < 0.05 or **P < 0.01
vs. TOPA. The “TOPA + BDNF (10)” and “TOPA + BDNF (50)” groups were not significantly
different from each other or from the no-treatment group (CTRL). Reprinted with permis-
sion from Wiley-Blackwell (15).
Table 1
Effect of BDNF on [3H]dopamine and [14C]GABA uptake
in control and TOPA-treated mesencephalic cells
5 None 50 ± 9 76 ± 11
BDNF 63 ± 14 86 ± 18
TOPA 6.9 ± 2.6* 9.3 ± 3.4*
BDNF/TOPA 43 ± 7** 15 ± 4
Mesencephalic cells were plated (5 × 105) in 25 mm diameter cluster wells (24-well
plate) and treated with 50 ng/mL BDNF on day 1. At 7 days, TOPA, where indicated,
was added to a final concentration of 10 μM. Twenty-four hours later, specific uptake
of [3H]dopamine and [14C]GABA was determined
*P < 0.01 vs. control (“none”) or BDNF-treated cultures; **P < 0.01 vs. TOPA-treated
cultures (n = 6). Reprinted with permission from Wiley-Blackwell (15)
9 Culture and Characterization of Rat Mesencephalic Dopaminergic Neurons 99
4. Notes
Acknowledgments
References
their striatal target cells. Proc Natl Acad Sci 13. Olney JW, Zorumski CF, Stewart GR, Price
USA 76, 5387–5391 MT, Wang G, and Labruyere J (1990) Excito-
10. Ferrari G, Minozzi M-C, Toffano G, Leon A, and toxicity of L-DOPA and 6-OH-DOPA:
Skaper SD (1989) Basic fibroblast growth factor Implications for Parkinson’s and Huntington’s
promotes the survival and development of mesen- diseases. Exp Neurol 108, 269–272
cephalic neurons in culture. Dev Biol 133, 140–147 14. Rosenberg PA, Loring R, Xie Y, Zaleskas V,
11. Qin L, Liu Y, Wang T, Wei SJ, Block ML, and Aizenman E (1991) 2,4,5-Trihydroxy-
Wilson B, et al (2004) NADPH oxidase medi- phenylalanine in solution forms a non N-methyl-
ates lipopolysaccharide-induced neurotoxicity D-aspartate glutamatergic agonist and neurotoxin.
and proinflammatory gene expression in acti- Proc Natl Acad Sci USA 88, 4865–4869
vated microglia. J Biol Chem 279, 1415–1421 15. Skaper SD, Negro A, Facci L, and Dal Toso R
12. Gao HM, Liu B, and Hong JS (2003) Critical (1993) Brain-derived neurotrophic factor
role for microglial NADPH oxidase in rote- selectively rescues mesencephalic dopaminergic
none-induced degeneration of dopaminergic neurons from 2,4,5-trihydroxyphenylalanine-
neurons. J Neurosci 23, 6181–6187 induced injury. J Neurosci Res 34, 478–487
Chapter 10
Abstract
Spinal cord motor neuron cultures are an important tool for the study of mechanisms involved in motor
neuron survival, degeneration and regeneration, volatile anesthetic-induced immobility, motor neuron
disorders such as amyotrophic lateral sclerosis or spinal muscular atrophy as well as in spinal cord injury.
Embryonic spinal cord motor neurons derived from rats have been successfully cultured; unfortunately,
the culture of adult motor neurons has been problematic due to their short-term survival. Recently, by
using a cocktail of target-derived factors, neurotrophins (brain-derived neurotrophic factor and glial cell
line-derived neurotrophic factor) and a permeable cyclic adenosine monophosphate analog, we have established
a reproducible protocol for long-term cultures of healthy and functional adult motor neurons (Exp Neurol
220:303–315, 2009). Here, we now describe in detail the steps that we used for the optimization of the
process of isolation and maintenance of adult rat ventral horn motor neurons in vitro.
Key words: Motor neurons, Spinal cord, Brain-derived neurotrophic factor, Glial cell line-derived
neurotrophic factor, Cyclic AMP, Conditioned medium, Adult rat
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_10, © Springer Science+Business Media, LLC 2012
103
104 J.V. Montoya-Gacharna et al.
Fig. 1. Flowchart protocol for the isolation and maintenance of adult rat spinal cord motor
neurons in vitro. Correlate the procedure with Fig. 2.
Fig. 2. Isolation and purification of spinal cord neurons. (a) Spinal cord obtained by hydraulic
spinal cord extrusion (modified from Meikle and Martin (34)). (b) Ventral horn segments in
neuronal isolation medium. (c) Density gradient for myelin removal (modified from Brewer (33)).
(d) Density gradient for neuron purification (modified from Brewer and Torricelli (29)).
106 J.V. Montoya-Gacharna et al.
Fig. 3. Adult rat spinal cord cultures 1 h (a) and 7 days (b) after plating. Cells were maintained with a cocktail of GDNF, BDNF,
cAMP, and C2C12 myocyte-conditioned medium (26). The arrows indicate large phase bright cells which are considered
morphologically as motor neurons. These neurons display extensive neurites in culture. Bar = 20 mm.
Fig. 4. Spinal cord neurons express a major synaptic protein, synaptophysin in vitro (7 days post plating). Cell cultures were
co-immunostained with microtubule-associated protein 2 (a) and synaptophysin (b). Synaptophysin puncta (arrows) are
found in the microtubule-associated protein 2-positive neurites. Inset : Neurites at higher magnification. Bar = 20 mm.
Fig. 5. KCl-evoked calcium cytoplasmic transients in dissociated adult rat spinal neurons (14 days in vitro). (a) The SCMNs
cultures were loaded with the calcium indicator Fura 2-AM (5 mM) (Molecular Probes) and stimulated with 100 mM KCl.
The excitation wavelengths for Fura (338 and 380 nm) were alternately (every 0.55 s) applied and the emitted fluorescence
was filtered with the fluorescence barrier filter BA 475 nm and collected with an intensified CCD camera (ATTO Instruments,
Rockville, MD, USA). (b) Phase bright light image of the spinal cord culture after KCl depolarization (40×).
10 Preparation of Adult Spinal Cord Motor Neuron Cultures… 107
2. Materials
2.1. C2C12 Myocyte- 1. C2C12 mouse myoblast cell line (CRL-1772, ATCC, VA).
Conditioned Medium C2C12 is a sub-clone (31) of the mouse myoblast cell line
established by Yaffe and Saxel (32).
2. C2C12 cell proliferation medium: Dulbecco’s modified Eagle’s
medium (DMEM), 10% fetal calf serum and 1% penicillin/
streptomycin. Store at 4°C.
3. C2C12 differentiation medium: DMEM, 1% penicillin/strep-
tomycin supplemented with 10% horse serum. Store at 4°C.
4. Cryoprotectant medium: DMEM, 10% fetal calf serum supple-
mented with 5% (v/v) dimethylsulfoxide.
5. 0.25% Trypsin-0.53 mM EDTA. Prepare 5 mL aliquots in
15-mL conical tubes. Store at −20°C.
2.2. Intracardiac 1. Adult Sprague–Dawley rats (70–100 days old) (see Note 1).
Animal Perfusion 2. A dose of 80 mg/kg ketamine (Ketaset® 100 mg/mL; Fort
Dodge Animal Health, IA) and 12 mg/kg xylazine (AnaSed®
20 mg/mL; Shenandoah, IA) is used per animal. Store anes-
thetics at room temperature in a locked cabinet.
3. Artificial cerebrospinal fluid solution (ACSF): 0.4 mM dex-
tran, 15 mM glucose, 3 mM KCl, 125 mM sucrose, 2.1 mM
HEPES, 1.2 mM KH2PO4, 125 mM glycerol, 1.3 mM MgSO4,
and 26 mM NaHCO3. Store at 4°C. Use within a month. On
the day of the spinal cord isolation, bubble the ACSF solution
for 45 min with 95% O2/5% CO2 on ice. Adjust the pH to 7.4
on ice with 1 N NaOH (see Note 2).
2.3. Spinal Cord Cell 1. Neuronal isolation medium (NIM): Hibernate A Medium
Isolation (Brain Bits, IL), 2% B27 supplement (Gibco, CA, 50×),
0.5 mM Glutamax, 100 U/mL penicillin, and 100 mg/mL
streptomycin (Gibco, CA) (see Note 3). Store at 4°C.
2. 2% DNAse I (Worthington, NJ) stock solution in NIM: Use
within 1–2 weeks. For long-term storage in solution follow the
manufacturer’s instructions. Store at −20°C.
3. Enzyme digestion solution (EDS): Prepare the day of neuron
isolation. Pre-warm 10 mL of NIM at 37°C and dissolve 20 mg
of papain (36 U/mL) (Worthington, NJ) (see Note 4). Add
108 J.V. Montoya-Gacharna et al.
2.4. Motor Neuron 1. Concentrated stock solutions of growth factors (made under
Feeding Medium sterile conditions): 1.1 mg/mL BDNF (Invitrogen, CA) and
0.4 mg/mL GDNF (Invitrogen, CA) in 0.1% bovine serum
albumin (manufacturer’s recommendation) (Sigma, MO) (see
Note 8). Store at −20°C for up to 6 months.
2. Concentrated stock solution of the cell-permeable cAMP analog:
25 mg/mL 8-(4 chlorophenylthio) cyclic adenosine-3¢,
5¢-monophosphate sodium salt (cAMP) (Sigma, MO) in dou-
ble distilled water. Store at −20°C.
3. Motor neuron feeding medium (MFM): Motor neuron basal
medium, 30% of C2C12 cell-conditioned medium, 1 ng/mL
BDNF, 0.1 ng/mL GDNF, and 125 mM cAMP.
10 Preparation of Adult Spinal Cord Motor Neuron Cultures… 109
3. Methods
3.2. Intracardiac 1. Set up the perfusion pump system (Masterflex®, Cole Parmer,
Animal Perfusion IL). Attach the perfusion needle and the tubing to the pump.
Run double distilled water through the tubing to remove any
residue. Fill up the tubing with cold ACSF and adjust the pump
to a slow steady drip (20–40 mL/min). The volume of ACSF
perfused is usually 200–300 mL per animal (see Note 10).
2. Anesthetize the rats with an intraperitoneal injection containing
a mixture of ketamine and xylazine following standard proto-
cols for anesthesia of small animals.
3. Test the level of anesthesia with a firm toe pinch. All surgical
procedures should be done under deep anesthesia.
4. Place the animal on its back. Palpate the sternum and the inferior
border of the last rib. These landmarks define the inferior
aspect of the rib cage. Make a transverse incision with sharp
scissors in the upper abdomen below the rib cage. Identify the
diaphragm and make a transverse incision to access the thoracic
cavity. Rapidly cut the ribs toward the head at the point of the
anterior thoracic line. Open up the thoracic cavity. Hold the
beating heart with forceps and insert the needle into the apex
of the heart. Clamp the inserted needle near the entry point.
Incise the right atrium and perfuse cold oxygenated ACSF
through the heart. A volume of approximately 200–300 mL of
ACSF is used per animal. The perfusion is completed when the
liver changes color from a dark red to a gray color, and the
extremities become white.
3.3. Spinal Cord Cell Spinal cord cells are isolated following the protocol by Brewer and
Isolation Torricelli (29) with some modifications.
1. Place the rat on its abdomen and decapitate it using a guillotine.
Incise the dorsal skin longitudinally from head to tail. Make a
transverse incision in the thigh to localize the femur. Fracture
both femurs with scissors as well as the spinal column at the level
of the sacrum, approximately 4 cm from the base of the rat’s tail.
Pull up the sacral vertebra and identify the spinal canal.
2. Place a Petri dish filled with cold NIM at the cervical/decapitated
end of the rat. Then, insert a syringe filled with 10 mL of
cold ACSF in the sacral canal and apply hydraulic pressure to
extrude the spinal cord into the NIM (Figs. 1 and 2a). Quickly
compress the plunger of the syringe and the spinal cord should
10 Preparation of Adult Spinal Cord Motor Neuron Cultures… 111
come out of the cervical end. Collect the spinal cord in cold
NIM (34) (see Note 11).
3. Immerse the spinal cord in the cold NIM (Fig. 2a) (see
Note 12).
4. The spinal cord should be immersed into a Petri dish containing
cold NIM at all times. Using a stereoscope, obtain transversal
sections of the spinal cord using a pair of iridectomy scissors
(Figs. 1 and 2b).
5. Tease away the meninges using small forceps.
6. Cut the spinal cord longitudinally through the posterior median
sulcus and the anterior median fissure to obtain two halves of
cord (each one containing a dorsal and a ventral column).
7. Identify a darker gray central area, which is butterfly shaped in
the cross-section of the spinal cord. The dorsal and ventral
horns are easily identified under a stereoscope. Separate the
dorsal and ventral columns by cutting longitudinally between
both ventral horns.
8. Cut the ventral horns in small pieces (2–3 mm) for better cell
dissociation.
9. Add 10 mL of EDS to a 15-mL conical tube. Transfer the ventral
horns to the conical tube and incubate for 5 min at room tem-
perature followed by 25 min at 30°C in a rotating incubator.
10. In the meantime, prepare 6 mL of 0.02% DNAse in EDS
medium by adding 60 mL of 2% DNAse to 6 mL of EDS
medium into a 15-mL conical tube.
11. Prepare three 2-mL fire-polished Pasteur pipettes (Fisher
brand) of decreasing diameter (large, medium, and small). The
Pasteur pipettes are prepared by flaming the pipette tips using
an alcohol burner.
12. Centrifuge the tissue at 1,228 × g for 5 min. All the following
steps (except for the centrifugation) should be done under a
sterile hood, and sterile techniques should be followed.
13. Remove the supernatant, and add 2 mL of NIM with 0.02%
DNAse to the pellet.
14. Triturate the tissue ten times with each of three Pasteur pipettes
starting with the pipette with the largest opening.
15. After each trituration, the homogenate is allowed to settle for
1 min and the supernatant is transferred into a separate 15-mL
conical tube.
16. Add an additional 2 mL of NIM to the precipitate and repeat
the process for a total of three times.
17. First gradient for myelin separation: Add 5 mL of 6% OptiPrep
1.32 to a 50-mL conical tube. Layer the final cell suspension
(6 mL) on top of the 6% OptiPrep 1.32 and centrifuge at
112 J.V. Montoya-Gacharna et al.
3.4. Feeding of Motor The cells are fed the day after plating, and every other day until the
Neuron Cultures end of the experiment. Care must be taken to minimize the time
the cells are outside of the incubator and exposed to ambient CO2
levels, as it can cause cell death. The cells should not be out of the
incubator for more than 3 min at a time.
To feed the cells:
1. Under the sterile hood, remove approximately 350 mL of
MFM (see Note 14).
2. Carefully add 350 mL of MFM by resting the pipette tip on
the side of the well to avoid detachment of the cells.
10 Preparation of Adult Spinal Cord Motor Neuron Cultures… 113
4. Notes
14. The SCMN cells are quite sensitive to temperature and pH,
and therefore it is essential to change the media quickly to
minimize the exposure to colder temperature and ambient pH
levels. If the cells remain outside for too long they are likely to
detach. We recommend the use of 2 × 2 Nunc® well plates
because they allow quick medium change. We also recommend
resting the well plate on a tube rack that is placed in the hood
(the night before, so it can be sterilized by the UV light)
because this reduces the contact the cells would have to the
relatively cold hood surface.
References
1. Zhang HM, Robinson N, Gomez-Curet I, 10. Carrascal L, Nieto-Gonzalez JL, Cameron WE,
Wang W, and Harrington MA (2009) Neuronal Torres B, and Nunez-Abades PA (2005)
and network activity in networks of cultured Changes during the postnatal development in
spinal motor neurons, Neuroreport 20, physiological and anatomical characteristics of
849–854 rat motoneurons studied in vitro, Brain Res
2. Streit J, Tscherter A, Heuschkel MO, and Brain Res Rev 49, 377–387
Renaud P (2001) The generation of rhythmic 11. Viana F, Bayliss DA, and Berger AJ (1994)
activity in dissociated cultures of rat spinal cord, Postnatal changes in rat hypoglossal motoneu-
Eur. J. Neurosci. 14, 191–202 ron membrane properties, Neuroscience 59,
3. Das M, Molnar P, Devaraj H, Poeta M, and 131–148
Hickman JJ (2003) Electrophysiological and 12. Berger AJ, Bayliss DA, Bellingham MC,
morphological characterization of rat embry- Umemiya M, and Viana F (1995) Postnatal
onic motoneurons in a defined system, Biotech. development of hypoglossal motoneuron
Prog. 19, 1756–1761 intrinsic properties, Adv Exp Med Biol 381,
4. Wallis DI, Elliott P, Foster GA, and Stringer 63–71
BM (1998) Synaptic activity, induced rhythmic 13. Nunez-Abades PA, and Cameron WE (1995)
discharge patterns, and receptor subtypes in Morphology of developing rat genioglossal
enriched primary cultures of embryonic rat motoneurons studied in vitro: relative changes
motoneurones, Can J Physiol Pharmacol 76, in diameter and surface area of somata and den-
347–359 drites, J Comp Neurol 353, 129–142
5. Fruns M, Krieger C, and Sears TA (1987) 14. Giller EL, Jr, Schrier BK, Shainberg A, Fisk
Identification and electrophysiological investi- HR, and Nelson PG (1973) Choline acetyl-
gations of embryonic mammalian motoneu- transferase activity is increased in combined
rones in culture, Neurosci Lett 83, 82–88 cultures of spinal cord and muscle cells from
6. Silani V, Braga M, Botturi A, Cardin V, Bez A, mice, Science 182, 588–589
Pizzuti A, and Scarlato G (2001) Human 15. Bennett MR, Lai K, and Nurcombe V (1980)
developing motor neurons as a tool to study Identification of embryonic motoneurons
ALS, Amyotroph Lateral Scler Other Motor in vitro: their survival is dependent on skeletal
Neuron Disord 2 Suppl 1, S69–76 muscle, Brain Res 190, 537–542
7. Bar PR (2000) Motor neuron disease in vitro: 16. Bennett MR, and Nurcombe V (1979) The
the use of cultured motor neurons to study survival and development of cholinergic neu-
amyotrophic lateral sclerosis, Eur. J Pharmacol rons in skeletal muscle conditioned media,
405, 285–295 Brain Res 173, 543–548
8. Jackson M, Ganel R, and Rothstein JD (2002) 17. Henderson CE, Huchet M, and Changeux
Models of amyotrophic lateral sclerosis, Curr JP (1984) Neurite-promoting activities for
Protoc Neurosci Chapter 9, Unit 9 13. embryonic spinal neurons and their develop-
9. Cameron WE, and Nunez-Abades PA (2000) mental changes in the chick, Dev Biol 104,
Physiological changes accompanying anatomi- 336–347
cal remodeling of mammalian motoneurons 18. Berg DK, and Fischbach GD (1978) Enrichment
during postnatal development, Brain Res Bull of spinal cord cell cultures with motoneurons, J
53, 523–527 Cell Biol 77, 83–98
116 J.V. Montoya-Gacharna et al.
19. O’Brien RJ, and Fischbach GD (1986) Isolation Muscle-conditioned media and cAMP
of embryonic chick motoneurons and their promote survival and neurite outgrowth of
survival in vitro, J Neurosci 6, 3265–3274 adult spinal cord motor neurons, Exp Neurol
20. Dohrmann U, Edgar D, Sendtner M, and 220, 303–315
Thoenen H (1986) Muscle-derived factors that 27. Das M, Bhargava N, Gregory C, Riedel L,
support survival and promote fiber outgrowth Molnar P, and Hickman JJ (2005) Adult rat
from embryonic chick spinal motor neurons in spinal cord culture on an organosilane surface
culture, Dev Biol 118, 209–221 in a novel serum-free medium, In vitro Cell
21. Smith RG, Vaca K, McManaman J, and Appel Dev Biol 41, 343–348
SH (1986) Selective effects of skeletal muscle 28. Das M, Bhargava N, Bhalkikar A, Kang JF, and
extract fractions on motoneuron development Hickman JJ (2008) Temporal neurotransmitter
in vitro, J Neurosci 6, 439–447 conditioning restores the functional activity of
22. Comella JX, Sanz-Rodriguez C, Aldea M, and adult spinal cord neurons in long-term culture,
Esquerda JE (1994) Skeletal muscle-derived Exp Neurol 209, 171–180
trophic factors prevent motoneurons from 29. Brewer GJ, and Torricelli JR (2007) Isolation
entering an active cell death program in vitro, J and culture of adult neurons and neurospheres,
Neurosci 14, 2674–2686 Nat Protoc 2, 1490–1498.
23. Arce V, Pollock RA, Philippe JM, Pennica D, 30. Tarsa L, and Goda Y (2002) Synaptophysin
Henderson CE, and deLapeyriere O (1998) regulates activity-dependent synapse formation
Synergistic effects of schwann- and muscle- in cultured hippocampal neurons, Proc Natl
derived factors on motoneuron survival involve Acad Sci USA 99, 1012–1016
GDNF and cardiotrophin-1 (CT-1), J Neurosci 31. Blau HM, Pavlath GK, Hardeman EC, Chiu
18, 1440–1448 CP, Silberstein L, Webster SG, Miller SC, and
24. Wiese S, Herrmann T, Drepper C, Jablonka S, Webster C (1985) Plasticity of the differenti-
Funk N, Klausmeyer A, Rogers M L, Rush R, ated state, Science 230, 758–766
and Sendtner M (2010) Isolation and enrich- 32. Yaffe D, and Saxel O (1977) Serial passaging
ment of embryonic mouse motoneurons from and differentiation of myogenic cells isolated
the lumbar spinal cord of individual mouse from dystrophic mouse muscle, Nature 270,
embryos, Nat Protoc 5, 31–38 725–727
25. Eagleson KL, and Bennett MR (1983) Survival 33. Brewer GJ (1997) Isolation and culture of
of purified motor neurones in vitro: effects of adult rat hippocampal neurons, J Neurosci
skeletal muscle-conditioned medium, Neurosci Meth 71, 143–155
Lett 38, 187–192 34. Meikle AD, and Martin AH (1981) A rapid
26. Montoya G J, Sutachan JJ, Chan WS, Sideris method for removal of the spinal cord, Stain
A, Blanck TJ, and Recio-Pinto E (2009) Technol 56, 235–237
Chapter 11
Abstract
Central neurons lose the ability for axonal regrowth during development and typically do not regenerate
their axons following axotomy once they become mature unless given a growth-permissive environment,
for example, a peripheral nerve graft. Retinal ganglion cells (RGCs) of the optic nerve represent a highly
useful cell model for the study of neurotrophic factor responsiveness, although the presence of nonneu-
ronal cells in the retina makes it difficult to interpret the direct effects of tested factors on RGCs. Cultures
of purified RGCs thus represent an excellent tool for the study of optic nerve cell trophic responsiveness,
in terms of both survival and axonal regeneration.
Key words: Retina, Ganglion cells, Rat, Neonatal, Embryonic, Culture, Antibody panning, Neurotrophic
factors, Axonal regeneration
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_11, © Springer Science+Business Media, LLC 2012
117
118 S.D. Skaper
2. Materials
2.1. Equipment 1. Stereo dissecting microscope with fiber optic light source.
and Labware 2. Laminar flow cabinet for dissections.
3. Laminar flow biological safety cabinet (CL2).
4. Humidified, water-jacketed culture incubator at 37°C and 5%
CO2/95% air.
5. Water bath set at 37°C.
6. Dissecting tools (Fine Science Tools).
7. Benchtop centrifuge to accommodate 15- and 50-mL tubes.
8. 15- and 50-mL polypropylene plastic centrifuge tubes (sterile).
9. 12-mL test tubes, sterile.
10. 6-mL flat bottom tubes (Bijou), sterile.
11. 1.5-mL Eppendorf tubes.
12. 10-cm ∅ sterile tissue culture dishes.
13. 6-, 10-, and 15-cm ∅ sterile Petri culture dishes.
14. #21- and #23-gauge hypodermic needles.
15. 5- and 10-mL syringes.
16. Nitex mesh, 20 μm (Tetko HC3-20).
17. 0.22- and 0.45-μm filters (Millipore).
18. Neubauer hemocytometer (Fisher Scientific).
19. 24- and 96-well tissue culture plates.
20. 24- and 96-well poly-D-lysine treated tissue culture plates (BD
Biosciences).
11 Rodent Retinal Ganglion Cell Cultures 119
2.3. Solutions for 1. Papain solution. Immediately before the start of the dissection,
Tissue Dissociation prepare a papain solution by adding 165 units (for postnatal day
8 (P8) rats, for embryonic, much less is needed) of papain to
10 mL of EBSS (or P-PBS or MEM) in a 15-mL blue-top centri-
fuge tube. Next, add 100 μL of a 4 mg/mL solution of DNase.
Dissolve the latter by placing the mixture, which is in a 15-mL
centrifuge tube, in a test tube rack in a 37°C water bath.
About 10 min before use, mix the papain solution with 2.4 mg
of L-cysteine hydrochloride and pass through a 0.22-μM filter
into a sterile scintillation vial or universal (Bijou) tube.
2. Ovomucoid solution. Dissolve 13 mg of ovomucoid and 13 mg
of BSA in 13 mL of MEM, and then add 0.13 mL of a 4 mg/mL
solution of DNase; adjust to pH 7.4 and filter-sterilize.
3. Trypsin (0.125%). Add 200 μL of a 2.5% trypsin stock to 4 mL
of CMF. Be sure to prepare the trypsin aliquots immediately
upon trypsin arrival; store at −20°C until use.
4. Trypsin inactivating solution. Combine 3 mL of FCS plus
10 mL DMEM.
5. Sato 100× stock solution. To 20 mL of Neurobasal medium, add
the following (final concentrations in brackets): 200 mg transferrin
(100 μg/mL), 200 mg BSA (100 μg/mL), 5 μL of stock pro-
gesterone solution (stock is 2.5 mg/100 μL ethanol) (60 ng/mL
or 0.2 μM), 32 mg putrescine (16 μg/mL), 200 μL of stock
sodium selenite (stock is 40 mg in 1 mL of 0.1 N NaOH plus
10 mL Neurobasal-A) (40 ng/mL), 10 μL of stock T3 (stock
is 8 mg in 1 mL of 0.1 N NaOH, 30 ng/mL), and 10 μL of
stock T4 (stock is 8 mg in 1 mL of 0.1 N NaOH, 40 ng/mL).
11 Rodent Retinal Ganglion Cell Cultures 121
Aliquot the above 100× stock solutions (200 μL) and store
at −20°C. The ethanol stocks for progesterone, sodium selenite,
T3, and T4 should be prepared fresh each time a new batch of
100× Sato medium is made.
3. Methods
the lens and vitreous humor with forceps. Then, gently lift the
retina away with a small spatula.
4. Store the retinas at room temperature in EBSS containing cal-
cium and magnesium, pH 7.4, until retinas are removed from
all animals.
3.2. Tissue 1. Upon completion of the dissection, remove the EBSS with a
Dissociation sterile Pasteur pipette and replace with 2 mL of the papain
solution (see Notes 3 and 4).
2. Transfer the retinas to the vial by pouring, and place the vial in
a test-tube rack in the water bath at 37°C. Every 10 min or so,
gently swirl the retinas. The incubation time with the papain
solution is 30 min (see Note 5).
3. Remove the vial from the water bath and transfer the retinas (by
pouring) to a 15-mL centrifuge tube. Allow the retinas to set-
tle, remove the papain solution with a pipette, and rinse the
tissue gently with 3 mL of ovomucoid solution. Allow the tis-
sue pieces to settle and remove the rinse solution (see Note 6).
4. Triturate the tissue with a 1-mL pipette in the ovomucoid
solution. The following steps are repeated ten times. Add
1 mL of ovomucoid solution, gently pull the tissue up into a
1-mL pipette and expel. Centrifuge the tissue dissociate for a
few seconds at 200 × g to remove pieces of tissue from the cell
suspension, and collect the supernatant. The retinal tissue is
further triturated by adding 1 mL of fresh ovomucoid solu-
tion, making one pass through the 1-mL plastic pipette, spin-
ning down the undissociated tissue, and collecting the
supernatant. After repeating this trituration procedure 6–10
times, the retinas should be completely broken up. A cell
count at this point should show a cell yield of about 20 million
cells/retina (see Note 7).
5. Centrifuge the resulting retinal cell suspension at 400 × g for
10 min to separate the retinal cells from the ovomucoid
solution.
6. Discard the supernatant and resuspend cells in 10 mL of MEM
containing 0.05% (w/v) BSA. The BSA serves to slow the cells’
settling time on the panning dishes. The cells are now ready for
the panning step.
3.3. RGC Purification 1. Incubate each of two 6-cm Petri dishes with 5 mL of 50 mM
by Panning Trizma buffer, pH 9.5, with affinity-purified goat anti-mouse
IgM, mu-chain specific (final concentration is 5–10 μg/mL)
3.3.1. Preparation
for 12 h at 4°C. To sterilize, pass the Tris solution through a
of the Panning Dish
filter into a sterile dish, and then add the azide-containing anti-
body directly to this; otherwise, much of the protein will be
lost to the filter (see Notes 8 and 9).
11 Rodent Retinal Ganglion Cell Cultures 123
3.3.3. Removing Adherent 1. Incubate adherent cells on the panning dish with 0.125%
Cells from the Panning trypsin solution for 10 min in a 37°C CO2 incubator.
Dish
2. Remove the dish from the incubator and squirt off the cells
with a 1-mL pipetman while they are still in the trypsin solu-
tion. Mark one point on the outside of the dish with marker.
Squirt from this point, once around the dish, by squirting from
the peripheral edge toward the center, rotate the dish, squirt
again, etc., until the entire surface area has been covered once.
Keep to a minimum the amount of squirting necessary to
remove the cells (see Note 11).
3. Transfer the cell suspension from the dish to a 15-mL centri-
fuge tube. Add 1 mL of the trypsin inactivating solution to
these cells and add another 4 mL to the dish. Again squirt the
dish. Check quickly under the microscope to make sure most
of the cells have been released (if not, this probably indicates
that the trypsin has lost activity).
4. Collect this 4 mL and transfer to the centrifuge tube (total
volume: 9 mL). Gently invert the tube several times to mix the
cells and remove a 100 μL aliquot for counting. Pour the
remainder of the trypsin inactivating solution into the tube
with the cells to top up the tube. Centrifuge at 200 × g for
5 min to collect the cells.
5. Resuspend the cells in Sato medium with growth factors for
purified RGCs, mix a 20 μL aliquot with an equal volume of
0.4% trypan blue, count, and plate. Expected yield: 40,000
RGCs viable (trypan blue–excluding cells) per P8 rat pup (see
Notes 12–15).
6. Culture the cells on a poly-D-lysine/merosin substrate. Dissolve
a 5-mg bottle of poly-D-lysine in 5 mL of 0.15 M borate buf-
fer, pH 8.4. Add 200 μL of this poly-D-lysine stock (1 mg/
mL) to 20 mL of tissue culture grade sterile water. Use 0.2 mL
poly-D-lysine per cm2 surface area. Incubate with poly-D-lysine
at room temperature (20–22°C) for 60 min. Merosin coating
is done after rinsing off excess poly-D-lysine: Add a 2 μg/mL
solution in Neurobasal medium and incubate at 37°C for sev-
eral hours to overnight (if overnight, add penicillin/strepto-
mycin) in a 5% CO2 incubator. The cells can be plated at 5,000
cells per well of a 96-well plate (high density) or at low density
on 6-cm tissue culture dishes, as desired.
3.4. Modifications 1. Use CD1 mice or other Thy1.2-positive strains, between 8 and
for Purifying Mouse P8 10 days postnatal age.
RGCs Rather than Rat 2. Instead of using an IgM Thy1.1 antibody, use an IgM Thy 1.2
antibody for the final panning dish. For example, the F7D5
Thy1.2 antibody, which is a mouse monoclonal (Accurate
Chemical Co., Cat No. MAS 731).
11 Rodent Retinal Ganglion Cell Cultures 125
3.5.3. Culture The same Neurobasal serum-free Sato medium, use of B27 at 1:50
is key, as well as BDNF, which will only save about a quarter of the
cells in any case. The E18 RGCs do not respond to the other
trophic factors.
3.6. MTT Survival 1. The MTT assay is frequently used to quantify neuronal cell
Assay survival. MTT reacts with mitochondrial dehydrogenases to
produce a blue formazan product in living cells, but not in
dying cells or their lytic debris (20, 21). Because of the low
density of RGC cultures, MTT-labeled cells are counted, rather
than dissolving the reaction product in dimethyl sulfoxide and
reading absorbance spectrophotometrically.
2. An MTT stock solution is prepared by dissolving MTT in
D-PBS at 5 mg/mL. Sterilize by passage through a 0.22-μm
filter. The stock solution can be stored at −20°C for up to
6 months.
3. Add 10 μL of the MTT stock to each 100-μL well of a 96-well
plate. Return the plate to the 37°C incubator for at least 1 h
(maximum 2 h). Viable cells with active mitochondria cleave
the tetrazolium ring into a visible dark blue/black formazan
reaction product (see Note 16).
4. Count the percentage of viable cells in each well by counting
at least 200 cells per well. Ideally, at least three wells per test
condition should be counted. Use an eyepiece reticle with a
counting grid. Cells must be counted immediately after the
MTT incubation to avoid the formation of large crystals of
MTT and toxicity of MTT to the cells.
126 S.D. Skaper
4. Notes
References
1. David S, and Aguayo AJ (1981) Axonal elon- in the CNS: implications for therapy. Regen
gation into peripheral nervous system “bridges” Med 3, 907–923
after central nervous system injury in adult rats. 3. Mansour-Robaey S, Clarke DB, Wang YC,
Science 214, 931–933 Bray GM, and Aguayo AJ (1994) Effects of
2. Gervasi NM, Kwok JC, and Fawcett JW (2008) ocular injury and administration of brain-
Role of extracellular factors in axon regeneration derived neurotrophic factor on survival and
11 Rodent Retinal Ganglion Cell Cultures 129
regrowth of axotomized retinal ganglion cells. 12. Unoki K, and LaVail MM (1994) Protection of
Proc Natl Acad Sci USA 91, 1,632–1,636 the rat retina from ischemic injury by brain-
4. Mey J, and Thanos S (1993) Intravitreal injec- derived neurotrophic factor, ciliary neu-
tions of neurotrophic factors support the sur- rotrophic factor, and basic fibroblast growth
vival of axotomized retinal ganglion cells in factor. Invest Ophthalmol Vis Sci 35,
adult rats in vivo. Brain Res 602, 304–317 907–915
5. Peinado-Ramon P, Salvador M, Villegas-Perez 13. Ma CHM, and Taylor JSH (2010) Trophic
MP, and Vidal-Sanz M (1996) Effects of axo- responsiveness of purified postnatal and adult
tomy and intraocular administration of NT-4, rat retinal ganglion cells. Cell Tissue Res 339,
NT-3, and brain-derived neurotrophic factor 297–310
on the survival of adult rat retinal ganglion 14. Barres BA, Silverstein BE, Corey DP, and Chun
cells. A quantitative in vivo study. Invest LL (1988) Immunological, morphological,
Ophthalmol Vis Sci 37, 489–500 and electrophysiological variation among reti-
6. Clarke DB, Bray GM, and Aguayo AJ (1998) nal ganglion cells purified by panning. Neuron
Prolonged administration of NT-4/5 fails to 1, 791–803
rescue most axotomized retinal ganglion cells 15. Zhang XM, Li Liu DT, Chiang SW, Choy KW,
in adult rats. Vision Res 38, 1,517–1,524 Pang CP, Lam DS, et al (2010) Immunopanning
7. Carmignoto G, Maffei L, Candeo P, Canella R, purification and long-term culture of human
and Comelli C (1989) Effect of NGF on the retinal ganglion cells. Mol Vis 16,
survival of rat retinal ganglion cells following 2,867–2,872
optic nerve section. J Neurosci 9, 1,263–1,272 16. Bader DR, MacLeish PR, and Schwartz EA
8. Lenzi L, Coassin M, Lambiase A, Bonini S, (1978) Responses to light of solitary rod pho-
Amendola T, and Aloe L (2005) Effect of toreceptors isolated from tiger salamander
exogenous administration of nerve growth fac- retina. Proc Natl Acad Sci USA 75,
tor in the retina of rats with inherited retinitis 3,507–3,511
pigmentosa. Vision Res 45, 1,491–1,500 17. Huettner JE, and Baughman RW (1986) Primary
9. Cui Q, Yip HK, Zhao RC, So KF, and Harvey culture of identified neurons from the visual cor-
AR (2003) Intraocular elevation of cyclic AMP tex of postnatal rats. J Neurosci 6, 3,044–3,060
potentiates ciliary neurotrophic factor-induced 18. Wysocki LJ, and Sato VL (1978) “Panning”
regeneration of adult rat retinal ganglion cell for lymphocytes: a method for cell selection.
axons. Mol Cell Neurosci 22, 49–61 Proc Natl Acad Sci USA 75, 2,844–2,848
10. Leaver SG, Cui Q, Plant GW, Arulpragasam A, 19. Brewer GJ, Torricelli JR, Evege EK, and Price
Hisheh S, Verhaagen J, et al (2006) AAV- PJ (1993) Optimized survival of hippocampal
mediated expression of CNTF promotes long- neurons in B27-supplemented Neurobasal, a
term survival and regeneration of adult rat new serum-free medium combination. J
retinal ganglion cells. Gene Ther 13, Neurosci Res 35, 567–576
1,328–1,341 20. Mosmann T (1983) Rapid colorimetric assay
11. Rios-Munoz W, Soto I, Duprey-Diaz MV, for cellular growth and survival: application to
Blagburn J, and Blanco RE (2005) Fibroblast proliferation and cytotoxicity assays. J Immunol
growth factor 2 applied to the optic nerve after Methods 65, 55–63
axotomy increases Bcl-2 and decreases Bax in 21. Manthorpe M, Fagnani R, Skaper SD, and
ganglion cells by activating the extracellular Varon S (1986) An automated colorimetric
signal-regulated kinase signaling pathway. J microassay for neuronotrophic factors. Dev
Neurochem 93, 1,422–1,433 Brain Res 25, 191–198
Chapter 12
Abstract
Glial cells play a key role in nervous system function, providing neurotrophic factor support to neurons as
well as taking part in two-way neuron-glia signaling (e.g., neurotransmitter release). White matter-derived
glia are important in certain neurodegenerative diseases involving axonal loss, for example in multiple scle-
rosis. Here we describe procedures for the preparation and culture of mixed nerve cells from postnatal rat
optic nerve, followed by protocols which can serve for the purification of individual populations of glia
from this tissue, namely O2A progenitors and oligodendrocytes, and astrocytes and astrocyte precursors.
Key words: Optic nerve, O2A progenitors, Oligodendrocytes, Astrocytes, Astrocyte precursors,
Immunopanning, Trophic factors, Differentiation, Cell culture
1. Introduction
Recent findings suggest that glial cells, though lacking the excitability
usually associated with most neurons, may be more actively involved
in brain function than has been previously thought. Collectively,
these findings indicate that glial cells can sense, and potentially
respond to, a large array of neuronal signals. Because glial cells are
intimately associated with most neurons, neurobiologists need to
consider the possible significance of active neuronal-glial signaling.
Although a variety of glial preparations have been studied, the
basic findings are common and are exemplified by studies of rat
optic nerve glia (1–3). Cultures of cells isolated from postnatal
optic nerves contain four types of macroglial cells that can be distin-
guished by their distinct antigenic phenotypes and morphologies:
type 1 astrocytes, type 2 astrocytes, oligodendrocytes, and a progeni-
tor cell, termed the O2A progenitor (4–6). The O2A progenitors
are developmentally bipotential, giving rise to both oligodendro-
cytes and type 2 astrocytes in vitro (7, 8). O2A progenitors persist
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_12, © Springer Science+Business Media, LLC 2012
131
132 S.D. Skaper
in adult animals (9, 10) in large numbers (11). These cell types are
not thought to comprise all kinds of central nervous system glia;
rather, they may be restricted to the kinds of glia present in white
matter. As such, they may be of considerable utility in the study of
white matter disease processes affecting the visual system. For
example, multiple sclerosis is characterized by optic nerve lesions
(12). This chapter details the preparation of mixed optic nerve cells
from postnatal or embryonic rat tissue, and serves as the “point of
departure” for subsequent methodology dealing with the purifica-
tion of oligodendrocytes, O2A progenitors, astrocytes, and astrocyte
precursor cells of optic nerve origin.
2. Materials
2.3. Stock Solutions 1. Low ovomucoid solution (10×). Dissolve 300 mg BSA and
300 mg ovomucoid in 20 mL D-PBS; adjust pH to 7.4. Pass
through 0.45-mm and then 0.22-mm filters attached to a 30-mL
syringe. Store in 1 mL aliquots at −20°C.
2. High ovomucoid solution (10×). Dissolve 2 g BSA and 2 g ovo-
mucoid in 20 mL D-PBS; adjust pH to 7.4. Pass through
0.45-mm and then 0.22-mm filters attached to a 30-mL syringe.
Store 0.6 mL aliquots at −20°C (see Note 3).
3. Forskolin (1,000×). Add 2.4 mL DMSO to a 10-mg bottle of
forskolin (=5 mM). Make 100 mL aliquots and store at −20°C.
The final concentration of solvent should not exceed 0.1% in
the culture medium.
4. N-acetyl cysteine (1,000×). Dissolve 50 mg N-acetyl cysteine in
10 mL of double-distilled water. Sterilize by passing through a
0.22-mm filter. Make 20 mL aliquots and store at −20°C.
5. IBMX (100 mM). Dissolve 22 mg IBMX in 10 mL D-PBS.
Sterilize by passing through a 0.22-mm filter. Make 40 mL aliquots
and store at −20°C.
6. Tris pH 9.5 (50 mM). Dissolve 12.1 g Trizma base in 200 mL
double-distilled water. Adjust pH to 9.5 with HCl.
7. Sato 100× stock solution. To 20 mL of Neurobasal-A medium
add the following (final concentrations in brackets): 200 mg
transferrin [100 mg/mL), 200 mg BSA [100 mg/mL], 5 mL of
12 Culture of Purified Glial Cell Populations from Optic Nerve 135
2.4. Sato Serum-Free 1. To 20 mL of Neurobasal-A medium add 200 mL Sato 100× stock
Culture Medium solution, 400 mL 50× B27 supplement, 200 mL sodium
pyruvate 100× stock, 200 mL glutamine 100× stock, 200 mL
of 100× penicillin/streptomycin stock (stored at −20°C),
20 mL N-acetyl cysteine 1,000× stock.
2. Next add 200 mL of 100× insulin stock (stored at 4°C; stock
must be prepared every 4–6 weeks—do not freeze). The 100×
insulin stock (0.5 mg/mL) is prepared by adding 2 mg of insulin
to 4 mL of tissue culture grade water containing 20 mL of 1 N
HCl. Filter the stock through a 0.22-mm filter and store at 4°C.
3. Filter the above medium through a 0.22-mm filter. Before fil-
tering the medium, filter about 5 mL of Neurobasal-A medium
to rinse away traces of detergent in the filter. The quality of the
filter matters—Millex-GV from Millipore is best. Falcon plastic
is preferable, along with Monoject syringes.
4. Recommended trophic factors for mixed optic nerve cultures:
Insulin (5 mg/mL) plus CNTF (10 ng/mL). Do not filter the
medium once the growth factors are added due to the risk of
their sticking to the filter. In addition, forskolin (5 mM) can be
included in the medium, as it promotes survival of mixed and
purified cultures.
3. Methods
3.1. Preparing an Optic 1. Prepare suspensions using papain only when they will be
Nerve Cell panned or when older nerves are being dissociated. The pro-
Suspsension Using cedures described here are modified from (12). Otherwise
Papain use the simpler trypsin/collagenase protocol (described
below—Subheading 3.2). Use sterile technique in all steps
(see Notes 4–6).
2. Dissect optic nerves and optic chiasm from postnatal day (P)
P6 to P10 rats (if you intend to collect oligodendrocytes use
P9–P10 animals to obtain a larger yield). Remove the head and
136 S.D. Skaper
skull and scoop the brain out—the optic nerves will be lying on
the base of the skull. Cut the nerves behind the chiasm and
also behind each eye and collect the nerves into a 35-mm Petri
dish containing 2 mL of HEPES-buffered MEM (pH 7.3–7.4)
(can also use D-PBS containing CaCl2 and MgCl2). Expect a
final yield of about 16,000 O2A cells per animal, and base your
rat usage on this (i.e., desired final number of cells).
3. Cut each nerve with a fine scissors or scalpel into as many pieces
as possible (at least 5–10 per nerve). Transfer the MEM (or
D-PBS containing CaCl2 and MgCl2) and nerve pieces to a
bijoux or polystyrene 12-mL test tube.
4. About 10 min before use, add 160 U of papain to 5 mL of
MEM/HEPES plus DNase stock 1:100 (stock is 0.4%) in a
bijou or polystyrene 12-mL test tube. Dissolve the papain by
placing this solution in a 37°C water bath for a few minutes.
Just before use, add L-cysteine (1 mg) to activate the papain
solution, adjust pH to 7.4 (as judged roughly by the color of
the phenol red pH indicator in the MEM, with 1 N NaOH).
Filter the final papain solution through a 0.22-mM filter. Be
sure to rinse the filter first with about 5 mL of MEM solution
to remove any traces of detergent in the filter.
5. Using a Pasteur pipette, remove the MEM/HEPES from the
vial containing the tissue and add the papain solution. Incubate
at 37°C for 75 min for P7 nerves, 90 min for P12 nerves, and
45 min for P0 nerves. Every 15 min gently agitate the tube.
6. Prepare 10 mL of the ovomucoid inhibitor solution: 9 mL of
MEM plus 1 mL of the “LOW” ovomucoid 10× stock solution
(containing 15 mg of BSA and 15 mg of ovomucoid per mL of
MEM) plus 1:100 (100 mL) of the DNase stock solution.
7. Use a Pasteur pipette to gently remove the papain solution.
Wash the optic nerve pieces gently by adding 1 mL of inhibitor
solution, allow the optic nerve pieces to settle, and then remove
the ovomucoid solution with a Pasteur pipette.
8. Serially triturate the nerve pieces in the ovomucoid inhibitor
solution, 1 mL at a time, with a 2.5-mL syringe and #21-gauge
needle × 2 passes, followed by a #23-gauge needle × up to 8
passes. That is, add 1 mL, pass the pieces through the needle 1
or 2 times, let the pieces settle, collect the suspension above
the pieces with a Pasteur pipette into a 10-mL test tube. Add
another mL of inhibitor solution, triturate again, and so on. In
this way, cells successively removed from the nerve pieces are
not repetitively passed through the needle (see Note 7).
9. If the optic nerve pieces fail to completely fall apart, it is likely
that enzyme concentration was too low; enzyme incubation
was too short; and enzyme lot was too old. Now, cap the tube
and gently invert once or twice to mix the cells.
12 Culture of Purified Glial Cell Populations from Optic Nerve 137
10. Filter the cell suspension through a 20-mm nylon mesh. Nitex
mesh from Tetko works well. Sterilize the mesh before use
(UV exposure for a few minutes is sufficient), prewet the mesh
on the bottom side, then form a cone with the filter on top of
a test tube, and pass 1 mL of MEM or inhibitor solution
through it before using a Pasteur pipette to transfer the cell
suspension through the filter.
11. Transfer a 100-mL sample to a 1.5-mL eppendorf tube for cell
counting (see Chapter 3).
12. Collect the cells by centrifuging at 200 × g for 10 min.
13. Gently remove the supernatant by aspiration. Using a 1-mL
Pipetman, resuspend the cell pellet in another ovomucoid solu-
tion containing 5.4 mL MEM plus 0.6 mL “High” ovomu-
coid 10× stock (contains 60 mg of BSA and 60 mg of
ovomucoid, but NO DNase). To resuspend, first add 1 mL of
this ovomucoid solution, resuspend with the 1-mL pipetman,
then add the remaining 5 mL of the ovomucoid solution, cap
tube, and gently invert once or twice to mix (see Note 8).
14. Centrifuge as in step 11, and aspirate supernatant. Use a 200-
mL pipetman to gently remove the residual solution, leaving
only the cell pellet.
15. Resuspend the cells in Sato serum-free medium at 106 cells per
mL. Yields are typically about 100,000 cells per pair of P7 optic
nerves.
16. Take a 100-mL aliquot for a cell count (see Chapter 3). Spin
down the rest of the cells and resuspend in about 7 mL of L15
or other air buffered solution containing insulin (0.5 mg/mL).
A 100× insulin stock is prepared by dissolving 2 mg of insulin
into 4 mL of H2O containing 20 mL of 1 N HCl (can be stored
at 4°C for 6 weeks). Insulin at high concentration activates
insulin-like growth factor-1 receptors on O2A progenitors and
oligodendrocytes (13), and is an essential survival factor for
O2A cells and oligodendrocytes (14, 15).
3.2. Preparing an Optic 1. Dissect and mince nerves in a 35-mm dish containing EBSS
Nerve Cell Suspension (see Note 9).
Using Trypsin and 2. Prepare enzyme solution containing 4 mL EBSS, 5 mg colla-
Collagenase genase, and 200 mL trypsin stock (stock is 2.5% in EBSS or
D-PBS and is stored at −70°C). Filter through a 0.22-mm filter.
Add DNase 1:100 from stock (stock is 0.4% in EBSS, stored at
−20°C).
3. Add nerves to trypsin solution in a bijou (or polystyrene 12-mL
test tube) and incubate for 20 min in a water bath at 37°C.
4. After this time add another 200 mL trypsin stock and incubate
again for 20 min. (incubate 20 min for P7 nerves and 10–15 min
138 S.D. Skaper
3.3. Adult Optic Nerve 1. Dissect (6–8 adult female rats) nerves into D-PBS, and cut
Dissociation each of the nerves into 15 pieces.
(Collagenase 2. Add collagenase (333 U/mL) in 4 mL of D-PBS and pass
and Trypsin) through a 0.22-mM filter. Incubate nerve pieces with this
solution at 37°C for 1 h.
3. Add 4 mL of trypsin (30,000 U/mL) in D-PBS; incubate at
37°C for 20 min.
4. Discard the trypsin solution and add 4 mL of 15,000 U/mL
trypsin in D-PBS with 0.27 mM EDTA; incubate at 37°C for
20 min.
5. Add an equal volume of 20% fetal calf serum in D-PBS (with
Ca2+, Mg2+) containing 0.08% DNase. Incubate at 37°C for
10 min.
6. Triturate with a 1-mL pipette (2×) before triturating with 21#
needle and 23# needle in low ovomucoid.
7. Wash in high ovomucoid as usual.
3.4. Preparation of 1. Dissociate the nerves with 0.125% trypsin in 4 mL D-PBS con-
Embryonic Optic Nerve taining 50 mL of 4% DNase stock.
Cells Using Trypsin 2. Incubate at 37°C for 30 min.
3. Triturate in 10% fetal calf serum buffer: (1 mL fetal calf serum,
9 mL D-PBS, 100 mL 4% DNase stock) using #21- and #23-
gauge needles as described above in Subheading 3.3.
4. Centrifuge at 200 × g for 10 min.
5. Resuspend in Sato serum-free medium and culture as desired.
3.5. Purification To purify O2A progenitors, an optic nerve cell suspension is pre-
Procedure for O2A pared by a papain dissociation procedure (modified from (16)) and
Progenitors passed sequentially over three antibody-coated “panning” (17)
dishes (papain must be used and not trypsin, to maximize cell yield
and to avoid destroying the RAN-2 surface antigen). First, a RAN-
2-coated dish is used which depletes the suspension of macrophages
(which stick to the first antibody-coated dish they are placed on),
meningeal cells, and type-1 astrocytes. In addition, about 20% of the
12 Culture of Purified Glial Cell Populations from Optic Nerve 139
3.6. Adult O2A Panning The panning procedure for adult O2As is very similar to that for
Procedure perinatal O2A progenitors. There are a few differences, as noted
12 Culture of Purified Glial Cell Populations from Optic Nerve 141
below. Apart from these, follow the original panning procedure for
perinatal O2A progenitors for the rest of the procedure. The number
of dishes and amounts of reagents are adequate for eight adult rats.
1. Prepare adult optic nerve suspension with collagenase and
trypsin (instead of papain).
2. Prepare 3–10-cm ∅ Petri plastic panning dishes: 2 RAN-2
dishes and 1 A2B5 dish (instead of RAN-2, galactocerebroside,
and A2B5 dishes): Add to each dish 12 mL of the Tris pH 9.5
buffer plus 40 mL of the affinity-purified antibody (anti-mouse
IgG + IgM antibody for the RAN-2 dish, and anti-mouse IgM
for the A2B5 dish).
3. Incubate the cell suspension on the panning dish as follows:
RAN-2 dish, 30 min; A2B5 dish, 45 min.
4. To remove the purified O2A cells from the A2B5 dish with
trypsin, use trypsin from Gibco (instead of Sigma).
5. To determine the cell yield, 10 mL (instead of 100 mL) of cell
suspension is directly dropped on a culture dish (three separate
drops for averaging). Let the cells settle down to the bottom of
the dish. Count the total cell number in the drop. Yield = num-
ber of cells in the drop × 100 × number of mL.
3.7. Purifying The simplest way is to let the purified O2A progenitors differenti-
Oligodendrocytes ate into oligodendrocytes by culturing for 2 days in survival factors
Instead of O2A (CNTF, insulin, forskolin) but not mitogens. To use the purified
Progenitors oligodendrocytes on the galactocerebroside dish, use O1 instead
of galactocerebroside antibodies to prepare this panning dish (O1
is an IgM, galactocerebroside is an IgG; it is easier to remove cells
from IgM-coated dishes).
3.8. Purification The following protocols are similar to the protocols for purifying
of Astrocytes and oligodendrocyte lineage cells. Only the modifications are described
Astrocyte Precursor below.
Cells from Rat Optic
Nerve
3.8.1. Astrocyte 1. Dissociate the minced optic nerves with 160 U papain in 5 mL
Purification from D-PBS with DNase and L-cysteine, and incubate at 37°C for
Perinatal Optic Nerves 45 min (P1), 60 min (P4), or 75 min (P8).
2. Triturate the tissue as for purification of O2A progenitors.
3. For panning, coat the first dish with a mouse monoclonal
MRC-OX7 anti-Thy1.1 antibody ((5 mg/mL) diluted in 4 mL
of PBS/BSA for a 10-cm ∅ dish) to deplete macrophages and
fibroblasts.
4. Coat the second dish with the mouse monoclonal anti-
A2B5 antibody (for P1). For purification from P4 or P8 optic
142 S.D. Skaper
3.9. Culturing Optic 1. Soak the desired quantity of 12-mm ∅ glass coverslips in 70%
Nerve Cells ethanol (prepared using MilliQ water) for at least 24 h prior to
use. Rinse with clean 70% ethanol and store in 70% ethanol
3.9.1. Coating Glass
until use.
Coverslips with
Poly-D-Lysine 2. Use sterile forceps to remove coverslips and place these lying
flat, but not overlapping each other, in a 10-cm ∅ Petri dish
containing autoclaved MilliQ water. Rinse several times with
this water to remove ethanol.
3. Add enough poly-D-lysine solution (dilute the 1 mg/mL poly-
D-lysine stock, which should be stored at −20°C, to 10 mg/mL
in autoclaved MilliQ water) to cover the slips (see Note 14).
4. Incubate the coverslips with poly-D-lysine for a minimum of
30–60 min.
5. Using a sterile forceps, individually pick up each coverslip, rinse
3 times with sterile MilliQ water (6-cm ∅ Petri dish works fine
for this), and place a single coverslip in each well of a 24-well
tissue culture plate.
6. Remove by aspiration any residual water in each well and allow
the coverslips to air dry in a laminar flow hood.
3.9.2. Plating Optic Nerve 1. Transfer 20 mL of the cell suspension (resuspended in Sato serum-
Cell Suspensions free medium, as described in preparation of the cell suspension)
onto the center of each poly-D-lysine-coated coverslip.
2. Incubate the multiwell plate with coverslips in a 5% CO2/95%
air incubator (37°C) for 15 min to allow the cells to adhere.
12 Culture of Purified Glial Cell Populations from Optic Nerve 143
4. Notes
References
1. Barres BA, Chun LLY, and Corey DP (1988) white matter glia: the type-l astrocyte. Neuron
Ion channel expression by white matter glia: I. 5, 527–544
Type 2 astrocytes and oligodendrocytes. Glia 4. Raff MC, Abney ER, Cohen J, Lindsay R, and
1, 10–30 Noble M (1983) Two types of astrocytes in
2. Barres B.A, Koroshetz WJ, Swartz KJ, Chun cultures of developing rat white matter: differ-
LLY, and Corey DP (1990) Ion channel ences in morphology, surface gangliosides, and
expression by white matter glia: the 02A glial growth characteristics. J Neurosci 3, 1289–l 300
progenitor cell. Neuron 4, 507–524 5. Miller RH, and Raff MC (1984) Fibrous and
3. Barres BA, Koroshetz WJ, Chun LLY, and protoplasmic astrocytes are biochemically and
Corey DP (1990) Ion channel expression by developmentally distinct. J Neurosci 4, 585–592
12 Culture of Purified Glial Cell Populations from Optic Nerve 145
6. Miller RH, ffrench-Constant C, and Raff MC 13. Zhao M, Wan ZL, Whittaker L, Xu B, Phillips
(1989) The macroglial cells of the rat optic NB, Katsoyannis PG, et al (2009) Design of an
nerve. Annu Rev Neurosci 12, 517–534 insulin analog with enhanced receptor binding
7. Raff MC, Miller RH, and Noble M (1983) A selectivity: rationale, structure, and therapeutic
glial progenitor cell that develops in vitro implications. J Biol Chem 284, 32178–32187
into an astrocyte or an oligodendrocyte 14. Pang Y, Zheng B, Fan LW, Rhodes PG, and
depending on culture medium. Nature 303, Cai Z (2007) IGF-1 protects oligodendrocyte
390–396 progenitors against TNF a-induced damage by
8. Raff MC, Williams BP, and Miller RH (1984) activation of PI3K/Akt and interruption of the
The in vitro differentiation of a bipotential glial mitochondrial apoptotic pathway. Glia 55,
progenitor cell. EMBO J 3, 1857–l864 1099–1107
9. ffrench-Constant C, and Raff MC (1986) The 15. Wilczak N, Chesik D, Hoekstra D, and De
oligodendrocyte-type 2 astrocyte cell lineage Keyser J (2008) IGF binding protein altera-
is specialized for myelination. Nature 323, tions on periplaque oligodendrocytes in multi-
335–338 ple sclerosis: implications for remyelination.
10. Wolswijk G, and Noble M (1989) Identification Neurochem Int 52, 1431–1435
of an adult-specific glial progenitor cell. 16. Huettner JE and Baughman RW (1986)
Development 105, 387–400 Primary culture of identified neurons from the
11. Barres BA (1991) New roles for glia. J Neurosci visual cortex of postnatal rats. J Neurosci 6,
11, 3685–3694 3044–3060
12. Green AJ, McQuaid S, Hauser SL, Allen IV, 17. Williams DL, Lo JL, and Amborski JF (1986)
and Lyness R (2010) Ocular pathology in mul- Enrichment of T lymphocytes from bovine periph-
tiple sclerosis: retinal atrophy and inflammation eral blood mononuclear cells using immuno-
irrespective of disease duration. Brain 133, affinity depletion technique (“panning”). Vet
1591–1601 Immunol Immunopathol 11, 199–204
Chapter 13
Abstract
In retinal diseases characterized by photoreceptor degeneration, the main cause of clinically significant
vision loss is cone, rather than rod, loss. Photoreceptor apoptosis can be affected by the availability and/
or delivery of neurotrophic proteins, and cultures of photoreceptors are valuable for studying these pro-
cesses. In the present study, a technique was designed to purify cones to make it possible to screen for
neuroprotective molecules. The present chapter describes a technique for preparing cultures of purified rat
retina cone photoreceptors based upon panning with peanut agglutinin lectin, which selectively binds to
cones. In addition, we describe a protocol for the purification and culture of retinal pigmented epithelial
cells from postnatal rat.
Key words: Cone photoreceptors, Retina, Pigmented epithelial cells, Rat, Cell culture, Neurotrophic
factors
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_13, © Springer Science+Business Media, LLC 2012
147
148 S.D. Skaper
2. Materials
22. L-Cysteine.
23. Insulin, from bovine pancreas (Sigma).
24. Transferrin, human.
25. Progesterone.
26. Putrescine.
27. Sodium selenite.
28. T3 (3,3¢,5-triiodo-L-thyronine, sodium salt), cell culture–
tested (Sigma).
29. Sodium pyruvate (100 mM stock), sterile, for cell culture (Sigma).
30. L-Glutamine (200 mM stock), sterile, for cell culture
(Invitrogen).
31. BDNF, human, recombinant (Peprotech).
32. CNTF, human, recombinant (CNTF) (Peprotech).
33. Forskolin.
34. N-acetyl cysteine.
35. Phenol red (0.5%), sterile, for cell culture (Sigma).
2.3. Stock Solutions 1. 10× EBSS stock solution without Ca2+, Mg2+, and bicarbonate.
1.16 M NaCl (68 g/L), 54 mM KCl (4 g/L), 10 mM
NaH2PO4·H2O (1.4 g/L), 10 mM D-glucose (10 g/L), and
0.05% phenol red (10 mL/L of 0.5% solution). Filter-sterilize
and store at 4°C.
2. D-Glucose, 30% (w/v). Dissolve 15 g of D-glucose in 50 mL
double-distilled water.
3. NaHCO3, 1 M. Dissolve 4.2 g of NaHCO3 in 50 mL double-
distilled water.
4. MgSO4, 100 mM. Dissolve 1.23 g of MgSO4·7H2O in 50 mL
double-distilled water.
5. EDTA, 0.5 M. Dissolve 7.3 g of EDTA in 50 mL double-dis-
tilled water.
6. EBSS-papain solution with Mg2+, EDTA, and bicarbonate.
Combine 20 mL of 10× EBSS solution, 2 mL of 100 mM
MgSO4, 2.4 mL of 30% D-glucose, 0.4 mL of 0.5 M EDTA,
and 5.2 mL of 1 M NaHCO3. Bring to a final volume of
200 mL with double-distilled water.
7. Borate buffer 0.15 M, pH 8.4. Dissolve 28.6 g of sodium borate
(Na2B4O7·10H2O) in 500 mL double-distilled water (pH will
be ~9.2). Adjust pH to 8.4 with 5 N HCl. Filter-sterilize and
store at 4°C (up to 6 months).
8. Low ovomucoid solution. Mix together 9 mL DPBS, 1 mL 10×
low ovomucoid stock (15 mg each BSA and ovomucoid/mL
DPBS), and 200 μL 0.4% DNase stock (made by dissolving
13 Isolation and Culture of Rat Cone Photoreceptor Cells 151
3. Methods
3.1. Cone 1. Rinse PNA dish 4 times with DPBS (mark side to pour) and
Photoreceptors rinse very well. Store in DPBS until ready to use (in the cold).
3.1.1. Complete 2. Add 1 mL OX-7 + 0.2 mL 4% BSA (40 mg/mL DPBS) + 3 mL
Preparation of Antibody DPBS to a 10-cm ∅ OX-7 dish.
Dishes (Day of Experiment) 3. Incubate the OX-7 dish at room temperature for 2 h on a flat
surface.
4. Rinse the OX-7 dish 3 times with DPBS and keep in DPBS
until needed (in the cold).
3.1.2. Equilibrate EBSS 1. Aliquot 4 mL EBSS (lacking calcium and magnesium) into one
and Sterilize Mesh Filter universal 10-mL tube (bijou) and 6 mL EBSS into another
tube.
2. Leave them to equilibrate in the 37°C 5%CO2/95% air incuba-
tor by leaving caps on loose. These solutions should change
color.
3. Expose the nitex nylon mesh (70-μm pore size) filter to UV
light under the vertical flow culture hood 20 min per side.
3.1.3. Dissociation 1. Dissect out postnatal day P17 mouse or rat retinas. Store
of Retina minced retinas in 35-mm petri dish in DPBS.
2. Equilibrate EBSS-papain (1 mM EDTA) buffer with
5%CO2/95% O2 for 15 min in a 37°C water bath.
3. Dissolve 100 U papain in 5 mL preequilibrated papain buffer.
Add 30 μL of a 2.4-mg/mL L-cysteine solution made up in
DPBS. Sterilize with a 0.22-μm filter. Remove the DPBS and
add retinas to papain by pouring. Add 200 μL of 0.4% DNase
and place in the 37°C bath.
4. Digest tissue for 90 min; gently agitate the tissue every
15 min.
5. Add DNase when needed (cells get sticky and do not settle).
This usually requires 400 μL of 0.4% DNase added over the
90-min period.
6. While waiting, make the following solutions: Panning buffer
containing 15 mL DPBS, 15 μL 100× insulin stock (see high-
insulin Sato medium), and 75 μL of 4% BSA.
154 S.D. Skaper
3.2. Culture 1. Cut off anterior portion of eye (make sure to remove all of
of RPE Cells the iris). Remove the lens, vitreous, cornea, and retina.
3.2.1. RPE Dissociation 2. Remove the RPE by pulling the RPE/choroid out of the eye
socket with forceps and store in Ca2+/Mg2+—free DPBS.
Remove the optic nerve.
3. Equilibrate the EBSS-papain (1 mM EDTA) solution with 5%
CO2/95% O2 for 15 min in a 37°C incubator.
4. Dissolve 100 U of papain and 333 U/mL of collagenase in
5 mL preequilibrated papain buffer. Next, add 0.8 mg of
L-cysteine to this solution.
4. Notes
References
1. Sancho-Pelluz J, Arango-Gonzalez B, progressive photoreceptor degeneration in RCS
Kustermann S, Romero FJ, van Veen T, rats. Eur J Neurosci 22, 1057–1072
Zrenner E, et al (2008) Photoreceptor cell 8. Fontaine V, Kinkl N, Sahel J, Dreyfus H, Hicks
death mechanisms in inherited retinal degen- D (1998) Survival of purified rat photorecep-
eration. Mol Neurobiol 38, 253–269 tors in vitro is stimulated directly by fibroblast
2. LaVail MM, Yasumura D, Matthes MT, Lau- growth factor-2. J Neurosci 18, 9662–9672
Villacorta C, Unoki K., Sung C-H, et al (1998) 9. Traverso V, Kinkl N, Grimm L, Sahel J, Hicks
Protection of mouse photoreceptors by sur- D (2003) Basic fibroblast and epidermal
vival factors in retinal degenerations. Invest growth factors stimulate survival in adult
Ophthalmol Vis Sci 39, 592–602 porcine photoreceptor cell cultures. Invest
3. Gao H, Hollyfield JG (1992) Aging of the Ophthalmol Vis Sci 44, 4550–4558
human retina: differential loss of neurons and 10. Faktorovich EG, Steinberg RH, Yasumura D,
retinal pigment epithelial cells. Invest Matthes MT, LaVail MM (1990) Photoreceptor
Ophthalmol Vis Sci 33, 1–17 degeneration in inherited retinal dystrophy
4. Curcio CA, Allen KA, Kalina RE (1993) delayed by basic fibroblast growth factor.
Aging of the human photoreceptor mosaic: Nature 347, 83–86
evidence for selective vulnerability of rods in 11. Fuhrmann S, Kirsch M, Hofmann HD (1995)
central retina. Invest Ophthalmol Vis Sci 34, Ciliary neurotrophic factor promotes chick
3278–3296 photoreceptor development in vitro.
5. Karl MO, Reh TH (2010) Regenerative medi- Development 121, 2695–2706
cine for retinal diseases: activating endogenous 12. Kassen SC, Thummel R, Campochiaro LA,
repair mechanisms Early changes in synaptic Harding MJ, Bennett NA, Hyde DR (2009)
connectivity following progressive photorecep- CNTF induces photoreceptor neuroprotection
tor degeneration in RCS rats. Trends Mol Med and Müller glial cell proliferation through two
16, 193–202 different signaling pathways in the adult zebrafish
6. Wenzel A, Grimm C, Samardzija M, Remé CE retina. Exp Eye Res 2009 88, 1051–1064
(2005) Molecular mechanisms of light-induced 13. Caffé AR, Söderpalm AK, Holmqvist I, van
photoreceptor apoptosis and neuroprotection Veen T (2001) A combination of CNTF and
for retinal degeneration. Prog Retin Eye Res BDNF rescues rd photoreceptors but changes
24, 275–306 rod differentiation in the presence of RPE in
7. Cuenca N, Pinilla I, Sauvé Y, Lund R (2005) retinal explants. Invest Ophthalmol Vis Sci 42,
Early changes in synaptic connectivity following 275–282
158 S.D. Skaper
14. Chong NH, Alexander RA, Waters L, Barnett Inhibition of nuclear translocation of apopto-
KC, Bird AC, Luthert PJ (1999) Repeated sis-inducing factor is an essential mechanism of
injections of a ciliary neurotrophic factor ana- the neuroprotective activity of pigment epithe-
logue leading to long-term photoreceptor sur- lium-derived factor in a rat model of retinal
vival in hereditary retinal degeneration. Invest degeneration. Am J Pathol 173, 1326–1338
Ophthalmol Vis Sci 40, 1298–1305 18. Miyazaki M, Ikeda Y, Yonemitsu Y, Goto Y,
15. Cepeda IL, Flores J, Cornfeldt ML, O’Kusky Kohno R, Murakami Y, et al (2008) Synergistic
JR, Doudet DJ (2007) Human retinal pigment neuroprotective effect via simian lentiviral vec-
epithelial cell implants ameliorate motor defi- tor-mediated simultaneous gene transfer of
cits in two rat models of Parkinson disease. J human pigment epithelium-derived factor and
Neuropathol Exp Neurol 66, 576–584 human fibroblast growth factor-2 in rodent
16. Ming M, Li X, Fan X, Yang D, Li L, Sheng models of retinitis pigmentosa. J Gene Med
Chen S, et al (2009) Retinal pigment epithelial 10, 1273–1281
cells secrete neurotrophic factors and synthe- 19. Miyazaki M, Ikeda Y, Yonemitsu Y, Goto Y,
size dopamine: possible contribution to thera- Sakamoto T, Tabata T, et al (2003) Simian len-
peutic effects of RPE cell transplantation in tiviral vector-mediated retinal gene transfer of
Parkinson’s disease. J Trans Med 2009, 7:53 pigment epithelium-derived factor protects
doi:10.1186/1479-5876-7–53 retinal degeneration and electrical defect in
17. Murakami Y, Ikeda Y, Yonemitsu Y, Onimaru Royal College of Surgeons rats. Gene Ther 10
M, Nakagawa K, Kohno R, et al (2008) 1503–1511
Chapter 14
Abstract
Retinal pigment epithelium cells play a key role in maintaining the normal function of retina and can
express several neurotrophic factors, which support the neurosensory retina and may also provide trophic
signals to the host dopaminergic neurons. The following chapter describes a protocol for the purification
and culture of retinal pigmented epithelial cells from postnatal rat.
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_14, © Springer Science+Business Media, LLC 2012
159
160 S.D. Skaper
2. Materials
2.3. Stock Solutions 1. 10× EBSS stock solution without Ca2+, Mg2+, and bicarbonate.
1.16 M NaCl (68 g/L), 54 mM KCl (4 g/L), 10 mM
NaH2PO4·H20 (1.4 g/L), 10 mM D-glucose (10 g/L), and
0.05% phenol red (10 mL/L of 0.5% solution). Filter-sterilize
and store at 4°C.
162 S.D. Skaper
3. Methods
3.1. Dissociation 1. Cut off anterior portion of eye (make sure to remove all of the
of RPE iris). Remove the lens, vitreous, cornea, and retina.
2. Remove the RPE by pulling the RPE/choroid out of the eye
socket with forceps and store in Ca2+/Mg2+—free DPBS.
Remove the optic nerve.
3. Equilibrate the EBSS-papain (1 mM EDTA) solution with 5%
CO2/95% O2 for 15 min in a 37°C incubator.
4. Dissolve 100 U of papain and 333 U/mL of collagenase in
5 mL preequilibrated papain buffer. Next, add 0.8 mg of
L-cysteine to this solution.
3.2. Purification of RPE 1. Using a 15-mL polystyrene centrifuge tube, layer metrizamide
(6 mL) with 40% on the bottom.
14 Culture of Rat Retina Pigmented Epithelial Cells 165
4. Notes
References
Abstract
The cell signalling mechanisms underlying mammalian central nervous system axon growth and guidance
change during development, such that axons that establish appropriate connectivity in the embryo fail to
regenerate after injury to the adult nervous system. The growth cone turning assay has been used in
Xenopus neurons to elucidate mechanisms of axon guidance during development. Here, we describe how
we have adapted this assay for rat dorsal root ganglion neurons to study the influence of extracellular
secreted factors causing growth cone attraction and repulsion. Additionally, we describe how this method
can be combined with small interfering RNA and cDNA transfections to manipulate protein expression in
growth cones, and fluorescence resonance energy transfer to monitor the activity of signalling pathways in
live neurons. This assay provides the unique ability to manipulate and visualise the internal status of growth
cone signalling whilst challenged with extracellular chemotropic signalling molecules, and can be used to
develop strategies to promote axon regeneration in the mature mammalian central nervous system.
Key words: Axon guidance, Regeneration, Growth cone, Turning assay, Fluorescence resonance
energy transfer, Cell signalling, cAMP
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_15, © Springer Science+Business Media, LLC 2012
167
168 A.J. Murray et al.
2. Materials
2.1. Dorsal Root 1. Bottenstein and Sato’s fluid (BSF). Ham’s F12 culture medium
Ganglion Neuron (Invitrogen) supplemented with 16 μg/mL putrescine,
Culture 100 μg/mL transferrin, 0.3% bovine serum albumin (BSA),
100 U/mL penicillin/streptomycin, 0.06 μg/mL progesterone,
0.16 μg/mL sodium selenite, 20 μg/mL insulin (all Sigma-
Aldrich) and 0.1 μg/mL nerve growth factor (NGF) (Serotec)
(see Note 1).
2. Low-walled, glass bottom culture dishes (MatTek).
3. Poly-L-lysine (70,000–150,000 mol. weight) (PLL) (Sigma-
Aldrich) and appropriate growth substratum, for example
laminin (Invitrogen).
4. 15% BSA dissolved in Ham’s F12 and stored in 2 mL aliquots
in 15-mL centrifuge tubes (Sterilin) at −20°C.
5. 37°C cell culture incubator with 5% CO2/95% air and class I
tissue culture hood.
6. Haemocytometer for counting cell numbers.
7. Trypsin inhibitor/DNAse. Hank’s balanced salt solution
(HBSS; Invitrogen) containing 0.002% trypsin inhibitor,
0.025% DNAse and 1% BSA (all Sigma-Aldrich).
2.3. Growth Cone 1. Glass pipettes (Intracell) pulled on a Sutter Pipette Puller to
Turning Assays have a 1 μm bore width.
2. Picospritzer III with a TSS10 dual pulse stimulator (both
Intracell) connected to a compressed air supply and micropi-
pette holder held by a micromanipulator.
3. Inverted phase contrast microscope with heated stage and CO2
supply and camera, connected to a computer equipped with
live imaging software. If CO2 is not available, HEPES buffer
can be used as an alternative.
4. ImageJ software for analysis of growth cone turning, available
freely from http://rsbweb.nih.gov/ij/.
170 A.J. Murray et al.
2.4. FRET Analysis 1. cDNA constructs designed to encode particular signalling pro-
teins with appropriate FRET-paired fluorophores.
2. Leica AF6000LX imaging system or equivalent FRET-capture
imaging system.
3. Leica application software or equivalent for analysis of FRET
time-lapse experiments.
3. Methods
3.1. Preparing Growth 1. Prepare 10 μg/mL PLL in sterile distilled H2O and gently
Substrata pipette 500 μL of this solution onto each central glass well of
MatTek low-walled 50-mm petri dishes under aseptic condi-
tions and leave overnight.
2. The next day, remove the PLL and allow the dishes to dry
completely in a tissue culture hood (see Note 2).
3. Prepare a solution of 2 μg/mL L-laminin in F12 medium and
gently pipette 500 μL into the central well of each MatTek dish.
4. Place dishes in a 37°C incubator for at least 2 h, but ideally
overnight. Other extracellular matrix substrates, such as
fibronectin, may also be applied in this way.
15 Mammalian Growth Cone Turning Assays Identify Distinct Cell… 171
Fig. 1. (a) Growth rates of DRG neuron neurites in a sealed culture dish, exposed to a heated (37°C) atmosphere and
‘sealed’ with a layer of oil. Note that the oil layer allows similar levels of neurite outgrowth to that seen in a sealed tissue
culture dish. (b) Typical responses of DRG neuron growth cones to a gradient of NGF emitted from the micropipette. The
bias of neurites growing in the direction of the gradient indicates an attractive response. (c) Position of the axon/growth
cone (left ) compared to the micropipette (right ). (d) Image from (c) showing the angle of the growth cone and distance
from the micropipette.
3.2. Preparation Here, we describe the culture of dissociated adult DRG neurons,
of Dissociated DRG as we believe they represent a good model for studying axon regen-
Neurons for Growth eration. However, we have also used this assay with DRG neurons
Cone Turning Assays at other developmental stages to study intrinsic cell signalling
changes in developing neurons (5, 7), and the procedure can be
used to study other mammalian neuronal types (10):
1. Dissect the spinal column from a sacrificed rat and remove as
much connective tissue and muscle as possible, then use cuticle
nippers to remove dorsal vertebral plates to expose the spinal
cord from the dorsal side.
2. Locate the DRGs lateral to the spinal cord on either side at each
vertebral segment, carefully cut the dorsal roots and place the
DRGs in warm F12 culture medium in a 35-mm petri dish.
3. Trim the roots from the DRGs and incubate the ganglia in F12
medium containing 0.125% collagenase for 1 h at 37°C in a 5%
CO2/95% air incubator.
4. Transfer the ganglia/collagenase to a 15-mL centrifuge tube
containing 10 mL F12 medium to wash out the collagenase.
Remove all F12 solution leaving the ganglia behind and add
172 A.J. Murray et al.
3.3. Set-up of 1. Using a Sutter Pipette Puller, pull 10-cm borosilicate glass
Picospritzer and pipettes with filaments, O.D. 1.0 mm, I.D. 0.5 mm (Intracell),
Equipment for Growth so that they have a bore width of 1 μm (see Note 9).
Cone Turning Assays 2. Connect a compressed air supply to a Picospritzer III under
the control of a TSS10 dual pulse stimulator (or equivalent) to
deliver air pulses at a frequency of 2 Hz for 20 ms duration, at
a pressure of 3 psi and a delay of 0.1 ms.
3. Using Eppendorf Microloader pipette tips and a P20 Gilson
(or equivalent) pipette, fill the tip of the glass micropipette
with the reagent to be assayed (4 or 5 μL will suffice), at a
1,000-fold greater concentration to that required at the growth
cone (2), and connect to the Picospritzer pipette holder
attached to a micromanipulator.
15 Mammalian Growth Cone Turning Assays Identify Distinct Cell… 173
3.4. Analysis of 1. Using ImageJ (NIH), measure the start angle of growth cone
Growth Cone Turning with respect to the micropipette by drawing a line through the
centre of the growth cone from the most proximal segment
of the neurite (this angle should be ~45° to the horizontal
position of the micropipette, from either above or below the
horizontal; see Fig. 1c, d).
2. Repeat step 1 with the image from the end of the assay (30 or
60 min); measure the angle between the final position of the
growth cone, its original position and the original direction of
growth at time zero. A final position of the growth cone on the
pipette side of the original 45° direction of growth represents
growth cone attraction to the gradient, whilst a final position
on the other side of the original 45° direction of growth repre-
sents growth cone repulsion.
3. Measure the distance that the growth cone has advanced
during the assay. If a growth cone has not advanced at least
10 μm, we consider it unresponsive because filopodia can
174 A.J. Murray et al.
3.5. FRET Combined It is assumed that growth cone turning is achieved by biased expression
with an Applied of intracellular signalling molecules leading to unilateral assembly
Gradient or disassembly of the cytoskeleton. We can now directly measure
protein activity across a growth cone when challenged with a
molecular gradient by combining the turning assay with FRET.
When a gradient is applied, changes in the distribution of FRET
across the growth cone indicate whether the extracellular gradient
activates or inhibits intracellular signalling proteins, revealing an
asymmetrical distribution of activity.
1. Plate DRG neurons transfected with appropriate FRET cDNA
constructs onto MatTek dishes coated as described in
Subheading 3.1, but use 5–10 μg/mL laminin to enhance
numbers of adherent transfected cells.
2. Analyse cells 24–48 h post-transfection using a FRET-capturing
system (e.g. see Subheading 2.4) using an inverted-stage
microscope set-up as described in Subheading 2.3.
3. Ensure FRET system and filters are properly configured for the
FRET probes being used and that appropriate correction
images are taken. These include untransfected, donor only and
acceptor only controls to correct for cross-talk between fluo-
rescent channels. Using FRET analysis software, these can be
used to generate correction factors in line with the algorithm
used by Wouters et al. (11).
4. Care should also be taken to avoid bleaching of the fluoro-
phores during time-lapse imaging. Control experiments should
be conducted to optimise exposure times, fluorescent light
intensity and frequency of image capture.
5. Select a growth cone that is flat, spread and contains lamellipodia
and filopodial protrusions. Care must also be taken to choose
a growth cone emitting suitable levels of fluorescence. Rotate
the dish so that the growth cone is facing the side of the
stage with the micromanipulator, at a 45° angle.
6. Having completed set-up and bleaching control measurements,
time-lapse experiments should be conducted by capturing
consecutive images in three separate channels: the donor chan-
nel (donor fluorophore excitation wavelength and donor fluo-
rophore emission capture), the FRET channel (donor fluorophore
excitation wavelength and acceptor fluorophore emission
capture) and the emission channel (acceptor fluorophore exci-
tation wavelength and acceptor fluorophore emission capture).
These channels are combined by the software to create a
pseudocolour sensitised FRET emission channel (FRET SE),
15 Mammalian Growth Cone Turning Assays Identify Distinct Cell… 175
3.6. Analysis of FRET 1. Regions of interest (ROIs) are outlined within the growth
Distribution cone for both the start and end point images. One ROI is
drawn outlining 50% of the surface area of the growth cone on
the side facing the pipette and the other is drawn outlining
50% of the growth cone on the side further from the pipette,
with the junction at which they meet bisecting the central axis
of the growth cone in line with the neurite shaft immediately
proximal to the growth cone.
2. A ratiometric value for the distribution of FRET across the
growth cone is obtained for each image using the formula:
NEAR SIDE FRET SE value/FAR SIDE FRET SE value.
A value <1 suggests the protein is less active on the near side of
the growth cone, whilst a value of >1 suggests the protein is
more active on the near half of the growth, i.e. the side exposed
to the gradient.
3. The procedure should be repeated for at least 15 growth cones,
with values for t0 and t5 plotted on a cumulative distribution
graph (see Fig. 2). Statistical comparison can be performed
comparing t0 and t5 values; a significant mean increase at t5
indicates a near side bias of signalling protein activity caused by
the gradient, whereas a significant mean decrease at t5
indicates a far side bias. Over longer time periods, this bias in
distribution of signalling molecule activity can be related to
growth cone attraction or repulsion.
176 A.J. Murray et al.
Fig. 2. Detection of asymmetric signalling activity within growth cones exposed to a gradient of the cAMP agonist,
Sp-cAMPS. (a) Near/Far ratio of FRET emission (abscissa) indicating the distribution of B-Raf FRET activity across embry-
onic rat DRG growth cones plotted against the percentage of growth cones that display that ratio or less (ordinate axis) at
the beginning of the experiment (t0, dark circles) and 5 min after application of the gradient (t5, light circles), with each
point representing one growth cone. The mean ratio for each group is indicated by corresponding symbols above the
abscissa. High values represent higher B-Raf activity on the side closest to the pipette, and values less than 1 represent
higher B-Raf activity on the side further from the pipette. Sp-cAMPS clearly instigates higher B-Raf activity on the side of
the growth cone towards the gradient. (b, c) Grayscale pictures of pseudocoloured FRET images at t0 (b) and t5 (c), with
darker colours indicating low FRET and lighter colours indicating high FRET. Arrow in (c) indicates the direction of the
applied gradient. Scale bars represent 10 μm. *P < 0.05, student’s t test.
4. Notes
1. BSF without insulin and NGF is stable at 4°C for several weeks.
After supplementing medium with NGF/insulin, BSF can be
stored for approximately 2 weeks at 4°C.
2. The PLL solution should be completely dried from the well
before applying the growth substrate, as liquid PLL is toxic to
cells.
3. Be very careful not to damage the neurons during the tritura-
tion step. Generally, a few passes using the glass Pasteur pipette
and Gilson P1000 pipette are sufficient.
4. 15% BSA can be prepared in advance and stored at −20°C. It
must be thawed in a 37°C water bath and mixed carefully
before use so that the BSA is evenly distributed.
5. If you are planning to carry out growth cone turning assays on
untransfected neurons, re-suspend the neuron pellet in
100 μL trypsin inhibitor/DNAse, before quickly adding
900 μL of BSF medium and moving on to step 8.
6. The optimum concentration of siRNA or plasmid DNA needs
to be established for each study. The transfection stage should
be completed as quickly as possible, ideally in less than 2 min,
15 Mammalian Growth Cone Turning Assays Identify Distinct Cell… 177
References
6. Murray AJ, Peace AG, Shewan DA (2009) acid (HEPES) as a tissue culture buffer. Proc
cGMP promotes neurite outgrowth and growth Soc Exp Biol Med 130, 305–310
cone turning and improves axon regeneration 10. Erskine L, Reijntjes S, Pratt T, Denti L,
on spinal cord tissue in combination with Schwarz Q, Vieira JM, Alakakone B, Shewan
cAMP. Brain Res 1294, 12–21 D, Ruhrberg C (2011) VEGF signaling
7. Murray AJ, Tucker SJ, Shewan DA (2009) through neuropilin 1 guides commissural axon
cAMP-dependent axon guidance is distinctly crossing at the optic chiasm. Neuron 70,
regulated by Epac and protein kinase A. J 951–965
Neurosci 29, 15434–15444 11. Wouters FS, Verveer PJ, Bastiaens PIH (2001)
8. Golding J, Shewan D, Cohen J (1997) Imaging biochemistry inside cells. Trends Cell
Maturation of the mammalian dorsal root entry Biol 11, 203–211
zone—from entry to no entry. Trends Neurosci 12. Tamada A, Kawase S, Murakami F, Kamiguchi
20, 303–308 H (2010). Autonomous right screw rotation
9. Shipman C Jr (1969) Evaluation of of growth cone filopodia drives neurite turning.
4-(2-hydroxyethyl)-1-piperazineethanesulfonic J Cell Biol 188, 429–441
Chapter 16
Abstract
The development of new therapeutics for management of pain is likely to become much more mechanism
based, and therefore, we need a more thorough understanding of the different pain development path-
ways. The afferent fibers of sensory neurons, with their cell bodies in the dorsal root ganglia (DRG), are
thought to be key in pain mechanisms. DRG neurons can be prepared from embryonic, postnatal, or adult
tissue. Embryonic preparations have the advantage of higher cell yields and greater proportion of neurons,
but they are dependent on neurotrophins for the first week of culture. In contrast, dissociated postnatal
and adult DRG sensory neurons offer the possibility to study mature neurons that may better resemble the
in vivo characteristics of these cells. Here, we describe the dissociation of DRG sensory neurons from
postnatal and adult rats. DRG are dissected and dissociated using a prolonged trypsin/collagenase treat-
ment, followed by mechanical separation of the neurons. We have routinely prepared them for electro-
physiological studies by the methods outlined in this chapter and describe some of the pitfalls that we have
encountered, with hints of how to overcome them.
Key words: Dorsal root ganglia, Dissociated cell culture, Growth factors, Rat, Sensory neurons,
Adult, Postnatal
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_16, © Springer Science+Business Media, LLC 2012
179
180 D.E. Owen and J. Egerton
inflammatory pain, but the sensory neurons are often affected early
and more severely. They are a key component of the nociceptive
pathway, and as such, we need to have a better understanding of
their role, in their response to and generation of inflammatory
mediators, and the control of their excitability in the development
of chronic pain.
The use of dissociated neurons, in contrast to acute prepara-
tions or slice cultures, means that a defined mechanism can be
more easily studied. The isolated neuron’s environment can easily
be manipulated in vitro to try and reflect the in vivo environment
or even the disease state. DRG neurons are very accessible and easy
to harvest in embryonic, young, and adult animals, and they survive
well in culture. Typically, DRG neurons are cultured from rats,
mice, or chicken embryos, and in most cases, one can obtain many
neurons from a single animal. For example, a complete preparation
from an adult rat yields ~40 DRGs or ~100,000 neurons. The yield
from an average age pup is ~500,000 neurons.
Postnatal DRG cultures can be used as a screening tool as the
yields are considerably greater and they grow well on multiwell
plate formats, which makes them open to fluorometric imaging
plate reader/calcium imaging (1), neurite outgrowth, cell health
assays, and cocultures. Unlike embryonic DRG neurons, postnatal
and adult DRG neurons do not need nerve growth factor (NGF)
or serum for survival (2), although the presence of NGF does affect
their phenotype. For example, NGF regulates the capsaicin sensi-
tivity of adult DRG neurons (3). The protocol in this chapter,
which involves dissociation of rat DRGs using a prolonged trypsin/
collagenase treatment, followed by mechanical separation of the
neurons, has been modified from Lindsay (2) and adapted for opti-
mum electrophysiological experimentation.
2. Materials
2.2. Adult DRG 1. Nutrient Mixture F-12 (with phenol red; Invitrogen).
Neurons 2. Ultroser G (Biosepra); reconstitute with 20 mL distilled water,
filter-sterilize, and add fresh to culture medium to 4% w/v.
3. Poly-DL-ornithine hydrobromide (Sigma P8638): Prepare a
25 mg/mL stock solution in distilled water and filter-sterilize.
Next, prepare a working solution of 500 μg/mL poly-DL-ornithine
by adding 200 μL of the 25 mg/mL stock to 10 mL of sterile
water.
4. Collagenase (Type IV, Sigma): Prepare a 6.25 mg/mL stock
solution (0.625% w/v) in F-12 medium (no phenol red) by
dissolving 50 mg in 8 mL medium and filter-sterilize. Further
dilute the collagenase to 0.625 mg/mL (0.0625% w/v) by
adding 200 μL of the 6.25 mg/mL stock to 1.8 mL of F-12
medium.
182 D.E. Owen and J. Egerton
3. Methods
3.1. Postnatal DRG 1. Sterilize dissection instruments by treating with 70% v/v etha-
Neurons nol and allow to dry in the dissection cabinet.
3.1.1. Dissection 2. Collect the DRG from 7- to 9-day-old rat pups (Sprague
Dawley). Cull the rat pups following Schedule 1 (cervical
dislocation followed by decapitation) according to Home
Office protocols.
3. Remove the tail and make an incision through the skin
following the line of the spinal column from the head to the
tail. Peel the skin away and trim away the adipose tissue from
the shoulders.
16 Culture of Dissociated Sensory Neurons from Dorsal Root… 183
3.1.2. Dissociation 1. Incubate the ganglia in the trypsin-EDTA for 15–20 min at
37°C.
2. Add 5 mL DMEM (10% FCS) to the dish with ganglia and
transfer to a 15-mL tube. Leave the DRG to settle to the bot-
tom or centrifuge at 200 × g for 3 min.
3. Carefully remove the supernatant using a 5-mL Stripette (do
not use vacuum aspiration).
4. Add 0.1% collagenase solution (1 mL of 0.5% stock plus 4 mL
HBSS) to the ganglia and incubate for 90 min at 37°C.
5. Add 5 mL DMEM (10% FCS) and centrifuge at 200 × g for
3 min.
6. Carefully remove the supernatant using a 5-mL Stripette (do
not use vacuum aspiration).
7. Add 1 mL DMEM (10% FCS) and mechanically dissociate the
ganglia by triturating no more than 6–10 times using a 1-mL
pipette tip.
8. Dilute the cell suspension (point 7) to 10 mL with DMEM
(10% FCS) and plate onto a 100-mm ∅ tissue culture dish for 1 h
at 37°C. The nonneuronal cells and large-diameter DRG neurons
184 D.E. Owen and J. Egerton
13. Contaminating glia that may still be present in the cultures can
be reduced by adding the antimitotic cytosine β-D-arabinoside
to a final concentration of 1 μM after 24 h in culture. This is
conveniently done by preparing a stock solution of 1 mM cyto-
sine β-D-arabinoside in water, filter-sterilizing, and storing as
aliquots at −20°C. This can be added 1:1,000 to the cultures.
3.2. Adult DRG 1. Sterilize dissection instruments by treating with 70% v/v etha-
Neurons nol, and allow to dry in the dissection cabinet.
3.2.1. Dissection 2. Weigh the rats and then sacrifice by exposure to a rising con-
centration of CO2. Confirm the death of each animal by expos-
ing the heart and severing one of the main cardiac blood
vessels. Perform exsanguination by placing each rat on its side
16 Culture of Dissociated Sensory Neurons from Dorsal Root… 185
3.2.2. Dissociation 1. Add 200 μL of 6.25 mg/mL collagenase (0.0625% final con-
centration) to the medium containing the DRG and swirl gen-
tly to mix. Incubate the ganglia for 2–3 h at 37°C, 5% CO2.
2. Using a 5-mL Stripette, carefully transfer the ganglia to a 15-mL
conical-bottomed centrifuge tube and wash twice with 10 mL
prewarmed growth medium to remove the collagenase.
3. Remove the medium by aspiration and resuspend the ganglia
in 2 mL growth medium. Triturate the DRG approximately 20
times through a flame-polished Pasteur pipette (smallest hole
possible) to obtain a single cell suspension.
4. Slowly drip the cell suspension onto a cushion of 2 mL 15%
BSA in growth medium in a 15-mL centrifuge tube and centri-
fuge at 200 × g at room temperature for 10 min to remove
nonneuronal cells and debris.
5. Carefully remove the supernatant by aspiration until approximately
50 μL remains covering the cell pellet. Resuspend the cell pellet
186 D.E. Owen and J. Egerton
4. Notes
References
1. Wood JN, Winter J, James IF, Rang HP, Yeats but are not required for survival of adult
J, and Bevan S (1988) Capsaicin-induced sensory neurones. J Neurosci 8, 2394–2405
ion fluxes in dorsal root ganglion cells in culture. 3. Winter J, Forbes CA, Sternberg J, and Lindsay
J Neurosci 8, 3208–3220 RM (1988) Nerve growth factor (NGF) regu-
2. Lindsay RM (1988) Nerve growth factors lates adult rat cultured dorsal root ganglion neu-
(NGF, BDNF) enhance axonal regeneration ron responses to capsaicin. Neuron 1, 973–981
Chapter 17
Abstract
This chapter presents fast and easy protocols to obtain highly purified cultures of proliferating adult rat,
canine, and human Schwann cells. Cell preparation from predegenerated adult sciatic nerves combined
with the use of melanocyte growth medium supplemented with forskolin, fibroblast growth factor-2, pituitary
extract, and heregulin as selective, serum-free culture medium and two methods for a consecutive cell-
enrichment step are described. Our protocols result in approximately 90% pure Schwann cell cultures (or
higher). The average time to obtain highly purified in vitro cultures of adult Schwann cells is 21 days.
Key words: Adult schwann cells, Rat, Human, Dog, Enrichment, In vitro
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_17, © Springer Science+Business Media, LLC 2012
189
190 K. Haastert-Talini
resulted in a reduction of the cell yield for adult rat and human
Schwann cell cultures, it was more effective than “cold jet” in the
purification of adult canine Schwann cells.
2. Materials
3. Methods
3.1. In Vitro 1. Harvest fresh peripheral nerve tissue and transfer it into
Predegeneration DMEM + 1% Pen/Strep (see Note 1).
2. Depending on the nerve anatomy, the procedure to remove
the epineurium with the help of Vannas microscissors and
17 Culture and Proliferation of Highly Purified Adult Schwann Cells… 193
3.2. Dissociation of For initial seeding of adult Schwann cells, the culture surfaces have
Predegenerated Tissue to be coated to enhance cell adherence.
and Initial Cell Seeding
3.2.1. Cell Culture Surface 1. Twenty-four hours before initial seeding, remove the PORN
for Adult Rat and Human stock solution from the +4°C storage and dilute 1:10 with ice-
Schwann Cell Cultures cold sterile distilled water (final concentration 1 mg PORN/
mL). Keep the PORN solution on ice and remove an aliquot
of laminin from −80°C and thaw it on ice.
2. Mix the ice-cold PORN solution with the laminin to give a
final concentration of 6-mg laminin/mL PORN.
3. For coating, cover the cell culture wells thoroughly (e.g.,
6-well plate: 1.5 mL/well) with ice-cold PORN-laminin solu-
tion and incubate for 24 h at room temperature (see Note 6).
4. Wash the wells with either distilled water or MGM and keep
them covered with either solution to avoid drying.
3.2.2. Cell Culture Surface Use PLL to coat plates for canine Schwann cell cultures.
for Adult Canine Schwann
1. Two hours before initial seeding, remove the prepared PLL
Cell Cultures
solution from the +4°C storage and thoroughly cover the cell
culture wells with it.
2. Incubate for 1 h, then wash the wells with either distilled water
or MGM and keep them covered with either solution to avoid
drying.
194 K. Haastert-Talini
3.2.4. Initial Seeding 1. Dilute the cell pellet in culture medium (see Note 8). To
enhance the seeding efficiency, add 1% BSA to the culture
medium for the first 24 h (see Note 9).
2. It is important to realize that seeding efficiency, initial survival
rate, and proliferation rate of primary adult Schwann cells
depend crucially on optimal cell densities in culture (26).
Therefore, count the number of viable cells prior to seeding,
e.g., after 1:2 dilution of a 10-mL cell suspension aliquot in
trypan blue (will stain dead/dying cells blue).
3. Plate primary adult peripheral nerve cells at a density of 106 living
(trypan blue-negative) cells/well on a 6-well plate (35 mm2).
4. The next day, change the medium to culture medium without
BSA and culture the primary peripheral nerve cells for 2–5 days
at +37°C and 5% CO2 (see Note 10).
3.3. Enrichment The fact that the adult Schwann cells grow mainly on top of the
of Adult Schwann Cell more flattened and more securely adherent fibroblasts allows
Cultures/Removal separation of the two cell types by an easy washing step.
of Fibroblasts 1. Remove the medium and slowly add 1 mL of ice-cold PBS and
3.3.1. Enrichment of Adult immediately aspirate for 2–3 times in order to flush the well
Rat and Human Schwann (6-well plate) and loosen the Schwann cells.
Cells 2. Apply a stream of 1-mL ice-cold culture medium, using a blue
1-mL Gilson tip and pipette on and off for several times,
detaching the Schwann cells from the underlying fibroblast
17 Culture and Proliferation of Highly Purified Adult Schwann Cells… 195
layer. Especially those parts of the wells with high cell densities
have to be rinsed. To reduce the risk of loosening also fibroblasts,
monitor the detachment using a phase-contrast microscope.
3. Transfer the suspension of floating cells, mainly Schwann cells,
to a 15-mL tube (see Note 11) and centrifuge (235 × g for
5 min at +21°C).
4. Resuspend the cell pellet in culture medium, determine the
viable cell number with trypan blue staining (see above), and
seed the enriched adult Schwann cells into freshly prepared
PORN-laminin-coated wells.
3.3.2. Removal of For enrichment of adult canine Schwann cells, “cold jet” does not
Fibroblasts from Adult work efficiently; however, these cells can be immunopurified.
Canine Schwann Cell
1. Wash magnetic Dynabeads® sheep anti-rat IgG in 0.1% BSA in
Cultures
PBS (10-mL beads in 500-mL 0.1% BSA). Initially, the
Dynabeads have to be fluffed up and then mixed with the
washing solution inside an eppendorf cup (1.5 mL).
2. Bring the magnet close to the wall of the cup; this draws the
magnetic Dynabeads toward the magnet, allowing the superna-
tant to be easily removed. Repeat the procedure 2 more times.
3. To couple the Dynabeads with rat anti-canine Thy1-antibody
(dilute 1:300 in MGM), add 10 mL of the washed Dynabeads
to 300 mL of the antibody solution in another eppendorf cup.
4. Securely seal the vial with parafilm, put it into an ice-filled
50-mL tube, and rotate it around its horizontal axis 45 min at
+4°C (e.g., place Roto-Shake Genie® in +4°C room). This
step facilitates equal coating of the beads with the antibodies.
5. Wash the antibody-coupled beads 3 times with 0.1% BSA as
before.
In parallel, all cells are enzymatically detached from the wells.
6. Remove the medium and incubate the cells with trypsin/
EDTA for 1 min at +37°C (within the CO 2 incubator),
followed by further incubation at room temperature and
observation under phase-contrast microscopy.
7. As soon as all cells are detached, add culture medium/collagenase
IV (dilute 1:1), pipette the cells on and off, and finally, transfer
them into a 15-mL tube for centrifugation later on.
8. Again, wash each well with 500-mL culture medium; add these
washes to the cell suspension.
9. Centrifuge at 235 × g for 5 min at +21°C.
10. Resuspend the cell pellet in culture medium/DNase I (dilute
10:1) and centrifuge again.
11. Resuspend the final cell pellet in culture medium (1 × 105
cells/300 mL).
196 K. Haastert-Talini
3.3.3. Conditions for After final purification of adult Schwann cells, the cultures can be
Propagation of Adult split whenever they reach confluency. For this, the cells are detached
Schwann Cell In Vitro with the use of trypsin/EDTA and reseeded on freshly prepared
PORN-laminin-coated culture dishes. Appropriate cell densities
for seeding enriched adult Schwann cells (purity > 95%) are as
follows:
● Adult rat Schwann cells: 5 × 104 cells/cm2.
● Adult canine and human Schwann cells: 2 × 105/cm2.
We do not include a proliferation step in our protocol as we
found proliferation rates to be enhanced in adult human Schwann
cells by permanently supplementing fresh heregulin to each
medium exchange (20). Elevated proliferation rates were induced
in adult rat Schwann cell cultures just by passaging the cells with
the “cold jet” technique (16). Adult canine Schwann cells show a
proliferation rate under the given conditions, which does not
require any further culture supplement.
Fig. 1. Highly purified human Schwann cell culture characterized by immunocytochemistry against the intracellular
calcium-binding protein S100 (a, b) or the cell surface molecule p75LNGFR (c). Nuclear counterstain with DAPI visualizes
S100- or p75LNGFR-immunonegative cells, e.g., fibroblasts, which additionally display darker and bigger nuclei in DAPI staining
(indicated by white arrows). Arrow heads in (a) indicate bright nuclei of dying cells.
4. Notes
Acknowledgments
In the first place, I wish to thank all persons who helped to estab-
lish and to advance the described techniques: Dr. Christina Mauritz,
Dipl. Biol.; Maike Wesemann; Sukhada Chaturvedi, PhD; Dr. Peer
Seef; Dr. Ruth Schmitte; and Silvana Taubeler-Gerling. Many
thanks also to Prof. Dr. Claudia Grothe for creating a supportive
working atmosphere. Special thanks go to our clinical colleagues
and their patients for providing us with human and canine nerve
tissue samples: Prof. Dr. Cordula Matthies; Prof. Dr. Götz Penkert;
Dr. Veronika Stein, PhD; Dr. Henning Schenk, PhD; Dr. Cornelia
Flieshardt; and Prof. Dr. Andrea Tipold.
References
1. Ansselin AD, Corbeil SD, Davey DF (1995) 7. Levi AD (1996) Characterization of the tech-
Culture of Schwann cells from adult animals. nique involved in isolating Schwann cells from
In Vitro Cell Dev Biol Anim 31, 253–254 adult human peripheral nerve. J Neurosci
2. Calderon-Martinez D, Garavito Z, Spinel C, Methods 68, 21–26
Hurtado H (2002) Schwann cell-enriched cul- 8. Keilhoff G, Fansa H, Schneider W, Wolf G
tures from adult human peripheral nerve: a (1999) In vivo predegeneration of peripheral
technique combining short enzymatic dissocia- nerves: an effective technique to obtain activated
tion and treatment with cytosine arabinoside Schwann cells for nerve conduits. J Neurosci
(Ara-C). J Neurosci Methods 114, 1–8 Methods 89, 17–24
3. Casella GT, Bunge RP, Wood PM (1996) 9. Keilhoff G, Fansa H, Smalla KH, Schneider W,
Improved method for harvesting human Wolf G (2000) Neuroma: a donor-age inde-
Schwann cells from mature peripheral nerve pendent source of human Schwann cells for tis-
and expansion in vitro. Glia 17, 327–338 sue engineered nerve grafts. Neuroreport 11,
4. Guenard V, Kleitman N, Morrissey TK, Bunge 3805–3809
RP, Aebischer P (1992) Syngeneic Schwann 10. Peulve P, Laquerriere A, Paresy M, Hemet J,
cells derived from adult nerves seeded in semi- Tadie M (1994) Establishment of adult rat
permeable guidance channels enhance periph- Schwann cell cultures: effect of b-FGF, alpha-
eral nerve regeneration. J Neurosci 12, MSH, NGF, PDGF, and TGF-beta on cell
3310–3320 cycle. Exp Cell Res 214, 543–550
5. Verdu E, Rodriguez FJ, Gudino-Cabrera G, 11. Komiyama T, Nakao Y, Toyama Y, Asou H,
Nieto-Sampedro M, Navarro X (2000) Vacanti CA, Vacanti MP (2003) A novel tech-
Expansion of adult Schwann cells from mouse nique to isolate adult Schwann cells for an
predegenerated peripheral nerves. J Neurosci artificial nerve conduit. J Neurosci Methods
Methods 99, 111–117 122, 195–200
6. Morrissey TK, Kleitman N, Bunge RP (1991) 12. Scarpini E, Kreider BQ, Lisak RP, Pleasure DE
Isolation and functional characterization of (1988) Establishment of Schwann cell cultures
Schwann cells derived from adult peripheral from adult rat peripheral nerves. Exp Neurol
nerve. J Neurosci 11, 2433–2442 102, 167–176
200 K. Haastert-Talini
13. Vroemen M, Weidner N (2003) Purification of nerve tissue engineering. J Neurosci Methods
Schwann cells by selection of p75 low affinity 186, 202–208
nerve growth factor receptor expressing cells 20. Haastert K, Mauritz C, Matthies C, Grothe C
from adult peripheral nerve. J Neurosci (2006) Autologous adult human Schwann cells
Methods 124, 135–143 genetically modified to provide alternative cel-
14. Pauls J, Nolte C, Forterre F, Brunnberg L lular transplants in peripheral nerve regenera-
(2004) [Cultivation and expansion of canine tion. J Neurosurg 104, 778–786
Schwann cells using reexplantation]. Berl 21. Bunge RP (1993) Expanding roles for the
Munch Tierarztl Wochenschr 117, 341–352 Schwann cell: ensheathment, myelination, tro-
15. Techangamsuwan S, Imbschweiler I, Kreutzer phism and regeneration. Curr Opin Neurobiol
R, Kreutzer M, Baumgartner W, Wewetzer K 3, 805–809
(2008) Similar behaviour and primate-like 22. Bunge RP (1994) The role of the Schwann cell
properties of adult canine Schwann cells and in trophic support and regeneration. J Neurol
olfactory ensheathing cells in long-term cul- 242(1 Suppl 1), S19–S21
ture. Brain Res 1240, 31–38 23. Stoll G, Jander S, Myers RR (2002)
16. Mauritz C, Grothe C, Haastert K (2004) Degeneration and regeneration of the periph-
Comparative study of cell culture and purifica- eral nervous system: from Augustus Waller’s
tion methods to obtain highly enriched cultures observations to neuroinflammation. J Peripher
of proliferating adult rat Schwann cells. J Nerv Syst 7, 13–27
Neurosci Res 77, 453–461 24. Jirsova K, Sodaar P, Mandys V, Bar PR (1997)
17. Haastert K, Mauritz C, Chaturvedi S, Grothe Cold jet: a method to obtain pure Schwann cell
C (2007) Human and rat adult Schwann cell cultures without the need for cytotoxic, apop-
cultures: fast and efficient enrichment and tosis-inducing drug treatment. J Neurosci
highly effective non-viral transfection protocol. Methods 78, 133–137
Nat Protoc 2, 99–104 25. Haastert K, Grosskreutz J, Jaeckel M, Laderer
18. Haastert K, Seef P, Stein VM, Tipold A, Grothe C, Bufler J, Grothe C, et al (2005) Rat embry-
C (2009) A new cell culture protocol for enrich- onic motoneurons in long-term co-culture
ment and genetic modification of adult canine with Schwann cells-a system to investigate
Schwann cells suitable for peripheral nerve tis- motoneuron diseases on a cellular level in vitro.
sue engineering. Res Vet Sci 87, 140–142 J Neurosci Methods 142, 275–284
19. Schmitte R, Tipold A, Stein VM, Schenk H, 26. Casella GT, Wieser R, Bunge RP, Margitich IS,
Flieshardt C, Grothe C, et al (2010) Genetically Katz J, Olson L, et al (2000) Density depen-
modified canine Schwann cells-In vitro and dent regulation of human Schwann cell prolif-
in vivo evaluation of their suitability for peripheral eration. Glia 30, 165–177
Chapter 18
Abstract
Cellular models composed of primary neuronal cultures or neuron-like cell lines are commonly used to
study neuron cell death and to test the neuroprotective properties of specific compounds. Cellular models
are easily accessible, permitting dissection and modulation of signaling pathways involved in neuron death.
For example, drug or shRNA delivery is more straightforward since there is no blood–brain barrier to
cross. However, since these models have their limitations, any important findings should ultimately be
verified with animal models and human samples. Here, we describe two cellular models that can be used
as a highly informative and easy to use starting point for testing potential neuroprotective drugs for
Parkinson’s disease: PC12 cells and sympathetic neuronal cell cultures. We describe in detail the protocols
needed to apply these models to study neuroprotection in the context of Parkinson’s disease.
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_18, © Springer Science+Business Media, LLC 2012
201
202 C. Malagelada Grau and L.A. Greene
2. Materials
All work should be carried out in a cell culture hood using sterile
technique. It is recommended to disinfect surfaces with 70% etha-
nol before and after the proceedings.
18 Use of PC12 Cells and Rat Superior Cervical Ganglion… 203
2.1. Coating Plastic 1. Rat tail collagen (Roche). Stock solution in 0.2% acetic acid in
Culture Ware sterile double distilled or deionized water. Working dilution
Substrates 1:15 in sterile double distilled or deionized water.
2.2. Coating Glass 1. Poly-D-lysine (Millipore) molecular weight >300 kDa. Dilute
Coverslip Substrates stock solution (1 mg/mL) in sterile double distilled water to
the final working concentration of 50 μg/mL.
2. Rat tail collagen (Roche). Stock solution in 0.2% acetic acid in
sterile double distilled or deionized water. Working dilution
1:15 in sterile double distilled or deionized water.
2.3. PC12 Cells 1. Complete medium: RPMI 1640 cell culture medium contain-
ing 10% heat inactivated horse serum (Sigma) (see Note 1), 5%
fetal bovine serum, and penicillin/streptomycin (50
units/50 μg of each per mL).
2. Differentiation medium: RPMI 1640 cell culture medium
containing 1% heat-inactivated horse serum and penicillin/
streptomycin (50 units/50 μg of each per mL) and 50 ng/mL
recombinant human or murine NGF (commercially available
from suppliers such as Alomone). This should be added from
1,000× stock just before use.
3. Freezing medium: complete medium with 10% dimethyl sul-
foxide (DMSO).
3. Methods
3.1. Growing PC12 To coat the plates, pipette the necessary amount of collagen (1:15)
Cells (see Note 2) to uniformly cover the surface. For a 10-cm culture dish, 1 mL is
sufficient, and this volume can be accordingly adjusted for culture
3.1.1. Coating Plasticware
dishes/wells of different sizes. Alternatively, one can add a larger
volume and then remove the excess with a sterile pipette. Allow the
plates to dry uncovered under a tissue culture hood for at least an
hour. After drying, plates can be stored at room temperature,
wrapped in aluminum foil or in the initial plastic bags, for 2–3
weeks (see Notes 3 and 4).
3.1.2. Thawing and Plating 1. If PC12 cells are received or stored in a frozen state (−80°C or
PC12 Cells in liquid nitrogen), they will be in complete medium and 10%
DMSO. Thaw the vial at 37°C in a water bath. This step should
be done quickly to diminish the toxicity of DMSO.
2. Prepare a sterile falcon tube with 10 mL of warm (37°C)
complete medium.
3. Add the thawed content of the cryotube to the 10 mL of
complete medium.
4. Mix well and centrifuge the cells for 5 min at 240 × g at room
temperature in a table top centrifuge.
5. Remove the supernatant (containing DMSO) and add 10 mL
of fresh complete medium to resuspend the cell pellet.
6. Mix well by trituration in a pipette and plate the contents in a
10-cm collagen-coated tissue culture plate. This step also serves
18 Use of PC12 Cells and Rat Superior Cervical Ganglion… 205
to break up cell clumps. Shake the plate well, back and forth
and left to right, to spread the cells uniformly. Avoid shaking
circularly or otherwise the cells will tend to concentrate at the
center of the plate.
7. Once the cells are plated, renew the culture medium every 2–3 days.
Approximately 2/3 of the medium should be exchanged.
Maintain a volume of 5–8 mL of medium.
3.4. Treating PC12 PC12 cells dramatically change their phenotype when they are
Cells with NGF exposed to NGF. They exit the cell cycle, project long neurite-like
processes, and take on many properties of differentiated sympathetic
neurons, including synthesis, storage, and release of catecholamines
(principally dopamine).
1. Detach PC12 cells from the plate as described in
Subheading 3.2.
2. Dilute the cell suspension with differentiating medium
(RPMI1640, 1% horse serum, and 50 ng/mL NGF). Typically,
206 C. Malagelada Grau and L.A. Greene
3.5. Cultivating From one rat litter of 12–14 pups, a 24- or 48-well plate can be
Sympathetic Neurons obtained. The cell density can be adjusted as desired by counting
from Rat Superior the cells at the end of the dissection. The steps below describe the
Cervical Ganglia preparation of cultures from one litter of newborn rat pups. A video
of the dissection and culture procedure can be found at (13).
1. Soak all the dissection tools in 70% ethanol for at least 10 min.
2. Spray or wipe down with 70% ethanol all the surfaces of the
dissecting hood, the microscope, and the fiber optic lights.
Also, disinfect with 70% ethanol the small polystyrene foam
support wrapped in aluminum foil. Put a sterile piece of gauze
over the support.
3. Prepare two 10-cm plates with 5 mL of RPMI or PBS. These
will be used to rinse the tools during the dissection to wash off
any adhering tissue.
4. Place the lids of the 10-cm plates face up in the hood and pipette
onto them one or two drops of RPMI for each pup that will be
dissected. After dissection, the ganglia will be transferred to
these drops for cleaning under the dissecting microscope.
5. Put the tubes with RPMI on ice.
6. Keep the pups in a clean box. Dissect one pup at a time.
7. Spray the pup for dissection with 70% ethanol and immediately
decapitate it using a pair of sharpened small dissecting scissors.
Remove the head closer to the shoulders than to the jaw so as
to avoid cutting through the ganglia.
8. Place the head on the polystyrene support facing up.
9. With the fine scissors, cut the skin of the neck. Pin the skin
flaps with the insulin needles to the polystyrene support. This
will hold the head in place and prevent the skin from folding
over the field of dissection.
10. Pipette RPMI onto the exposed tissue to remove blood cells
and debris and to prevent drying.
18 Use of PC12 Cells and Rat Superior Cervical Ganglion… 207
3.6. Toxin Treatments Both NGF-differentiated PC12 cells and sympathetic neurons will
be ready to use after at least 7 days following plating.
1. Prepare stock solutions of the toxins. Use stock concentrations
to minimize the volume of solution added to the cultures.
2. Change the medium right before any treatment.
3. If a specific compound is tested in conjunction with the toxins,
pretreatment should be done at least 30 min to 1 h before add-
ing the toxin. This is to permit entry into the cells. Always
include controls of sister cultures treated with the vehicles used
for delivery of toxin or tested agent. If the vehicle is DMSO,
the final amount should be no more than 0.1% of the final vol-
ume in the culture well.
4. To treat neuronal PC12 cells with (see Notes 9–12):
– 6-OHDA: a stock of 10 mM is recommended to treat the
cells at a final concentration of 50–100 μM.
– MPP+: a stock of 100 mM is recommended to treat the
cells at a final concentration of 500 μM–1 mM.
5. To treat rat sympathetic neurons, prepare stocks as above, but
the final concentrations of the toxins should be much lower:
5–10 μM for 6-OHDA and 50–100 μM for MPP+ (see Notes
9–12).
6. After treatments, wait for the desired time to assess the cul-
tures. If viability is monitored, 24 and/or 48 h are suggested.
7. For analyzing the samples by Western immunoblotting, choose
shorter incubation times.
3.7. Viability To quantify viable cells, there are several methods to choose from.
Assessment However, 6-OHDA and MPP+ with their capability to inhibit
mitochondrial complexes can interfere with tetrazolium salt assays,
leading to an overestimation of cell death. We routinely use a
method with a detergent solution that dissolves the cell plasma
membrane and leaves the nuclear membrane intact (17). This per-
mits counting of the numbers of surviving cells in the cultures.
With a Neubauer hemacytometer chamber, healthy round nuclei
can be counted under a phase microscope and distinguished from
the smaller, darker dead or degenerating nuclei.
The procedure is as follows:
1. Aspirate to fully remove the medium.
2. Add 0.5 mL of detergent solution to the wells (in a 48-well
plate, add accordingly for other-size dishes or wells) and mix
well (see Note 13).
3. Place approximately 10 μL of the nuclear suspension under the
coverslip of the Neubauer chamber.
4. Count at least 100 nuclei (see Note 14).
18 Use of PC12 Cells and Rat Superior Cervical Ganglion… 209
4. Notes
Acknowledgments
References
1. Greene LA, and Tischler AS (1976) mesencephalon-derived cells. Brain Res 866,
Establishment of a noradrenergic clonal line of 33–43
rat adrenal pheochromocytoma cells which 10. Malagelada C, and Greene LA. PC12 cells as a
respond to nerve growth factor. Proc Natl model for Parkinson’s disease research., in
Acad Sci USA 73, 2424–2428 Parkinson’s Disease: molecular and thera-
2. Greene LA (1978) Nerve growth factor pre- peutic insights from experimental models. ,
vents the death and stimulates the neuronal N. Przedborski, Editor. 2008, Elsevier.
differentiation of clonal PC12 pheochromocy- p. 375–389
toma cells in serum-free medium. J Cell Biol 11. Fahn S (1998) Medical treatment of Parkinson’s
78, 747–755 disease. J Neurol 245 Suppl 3, P15–24
3. Greene LA, and Rein G (1977) Release of 12. Marras C, and Lang A (2008) Invited article:
(3H)norepinephrine from a clonal line of pheo- changing concepts in Parkinson disease: mov-
chromocytoma cells (PC12) by nicotinic cho- ing beyond the decade of the brain. Neurology
linergic stimulation. Brain Res 138, 521–528 70, 1996–2003
4. Walkinshaw G, and Waters CM (1994) 13. Zareen N, and Greene LA (2009) Protocol for
Neurotoxin-induced cell death in neuronal culturing sympathetic neurons from rat supe-
PC12 cells is mediated by induction of apoptosis. rior cervical ganglia (SCG). J Vis Exp,
Neuroscience 63, 975–987 2009(23)
5. Nakamura K, Bindokas VP, Marks JD, Wright 14. Malagelada C, Ryu EJ, Biswas SC, Jackson-
DA, Frim DM, Miller RJ, et al (2000) The Lewis V, and Greene LA (2006) RTP801 is
selective toxicity of 1-methyl-4-phenylpyridinium elevated in Parkinson brain substantia nigral
to dopaminergic neurons: the role of mito- neurons and mediates death in cellular models
chondrial complex I and reactive oxygen species of Parkinson’s disease by a mechanism involv-
revisited. Mol Pharmacol 58, 271–278 ing mammalian target of rapamycin inactiva-
6. Hirata Y, and Nagatsu T (2005) Rotenone and tion. J Neurosci 26, 9996–10005
CCCP inhibit tyrosine hydroxylation in rat stri- 15. Schober A (2004) Classic toxin-induced ani-
atal tissue slices. Toxicology 216, 9–14 mal models of Parkinson’s disease: 6-OHDA
7. Sherer TB, Betarbet R, Testa CM, Seo BB, and MPTP. Cell Tissue Res 318, 215–224
Richardson JR, Kim JH, et al (2003) Mechanism 16. Ryu EJ, Harding HP, Angelastro JM, Vitolo
of toxicity in rotenone models of Parkinson’s OV, Ron D, and Greene LA (2002) Endoplasmic
disease. J Neurosci 23, 10756–10764 reticulum stress and the unfolded protein
8. Yang WL, and Sun AY (1998) Paraquat- response in cellular models of Parkinson’s dis-
induced cell death in PC12 cells. Neurochem ease. J Neurosci 22, 10690–10698
Res 23, 1387–1394 17. Rukenstein A, Rydel RE, and Greene LA
9. Zhou W, Hurlber MS, Schaack J, Prasad KN, (1991) Multiple agents rescue PC12 cells from
and Freed CR (2000) Overexpression of human serum-free cell death by translation- and tran-
alpha-synuclein causes dopamine neuron death scription-independent mechanisms. J Neurosci
in rat primary culture and immortalized 11, 2552–2563.
Chapter 19
Abstract
Neurotrophic factors released by target tissues maintain the survival and differentiation of innervating
neurons. The manner by which these target-derived neurotrophic proteins communicate with innervating
neurons has been actively pursued for over three decades. The present chapter describes a technique for
preparing and maintaining compartmented chambers for culturing neurons derived from either superior
cervical ganglia (sympathetic neurons) or dorsal root ganglia (sensory neurons). This system recapitulates
the selective stimulation of neuron terminals that occurs in vivo following release of target-derived
neurotrophins.
Key words: Campenot cultures, Superior cervical ganglia neurons, Dorsal root ganglia neurons, Cell
culture, Neurotrophic factors, Retrograde transport
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_19, © Springer Science+Business Media, LLC 2012
213
214 S.D. Skaper
side compartments
teflon divider
collagen scratches
center compartment
culture dish
Fig. 1. Schematic diagram of Campenot chamber culture setup for isolation of distal neuronal
axons. A 35-mm petri dish is divided into three chambers by a Teflon divider (7.5 mm high)
that is sealed to the floor of the dish (a collagen-coated coverslip) with silicone grease.
The floor of the narrow (1 × 5 mm) central compartment in which neurons are plated is
transected by a series of parallel scratches which guide the growing neurites into the left
and right side compartments. The lower portion of the diagram shows an illustrative
enlargement of the neurons on a single track. Compartmented cultures isolate cell bodies
and proximal axons and distal axons of neurons. See text for further details.
2. Materials
2.2. Compartmented 1. Teflon dividers (Camp 10, Tyler Research, Edmonton, Alberta,
Chamber Components Canada) (see Note 1)
2. Pin rake (Camp-PR, Tyler Research)
3. Grease loader (Camp-GLSS, Tyler Research)
4. Corning high-vacuum grease (Fisher Scientific)
5. 23-gauge Luer Lock connector (Fisher Scientific)
6. Right-angled hemostatic forceps (Fine Science Tools)
7. Collagen solution, type 1 from rat tail, for cell culture, sterile
filtered (Sigma-Aldrich)
8. 7.5% bovine serum albumin (BSA) solution (Invitrogen)
9. Penicillin/streptomycin, 5,000 U/mL penicillin + 5,000 μg/mL
streptomycin (100× stock), sterile, for cell culture (Invitrogen)
10. Cytosine β-D-arabinofuranoside (Ara-C) (Sigma-Aldrich)
11. N2-methylcellulose 400CPS (Xenex Laboratories, Coquitlam,
BC, Canada)
12. Brain-derived neurotrophic factor, recombinant human
(PeproTech)
13. NGF, recombinant human (PeproTech)
14. NOCHROMIX® (Sigma-Aldrich)
2.4. Culture Media 1. Plating medium for newborn rat DRG neurons: Supplement
and Other Solutions 50 mL of DMEM to contain N2 supplements (500 μL
from 100× stock), 0.05% BSA (334 μL from 7.5% BSA
solution), 50 U/mL penicillin, and 50 μg/mL streptomy-
cin (5 mL of 100× stock). NGF is added as appropriate
(see Subheading 3.2).
2. Plating medium for newborn rat SCG neurons: Supplement
50 mL of RPMI-1640 to contain 50 ng/mL NGF (add
50 μL from a 50 μg/mL stock), 1% heat-inactivated horse
serum, and 50 U/mL penicillin and 50 μg/mL streptomycin
(5 mL of 100× stock). NGF is added as appropriate (see
Subheading 3.2).
3. N2-methylcellulose: Weigh out 0.6 g of methylcellulose and
place it in a 250-mL bottle, add a stir bar, and autoclave it for
20 min on dry (from this point, all work must be sterile). Next,
add 200 mL of serum-free medium (RPMI-1640 or DMEM)
supplemented with N2 components and stir in a cold room
until it dissolves. Aliquot into 15-mL tubes and store at −20°C.
To facilitate routine use, prepare also 1-mL aliquots and store
at −20°C.
4. Ara-C is prepared as a 10-mM stock solution in phosphate-
buffered saline, filter-sterilized, and stored as aliquots at −20°C
for up to 6 months. Once thawed, the aliquot tube may be
kept at 4°C for 4 weeks. Where used, Ara-C is diluted in
culture medium to a final concentration of 1 μM.
2.5. Preparation 1. Dilute the stock collagen solution to 1 mg/mL using 0.001 N
of Collagen HCl as diluent (alternative, tissue culture grade water can be
Substratum used). Add 1.5 mL of the diluted collagen solution per 35-mm
tissue culture plate and leave overnight in the CO2 incubator at
37°C. Then, remove the dishes from the incubator, aspirate to
collagen solution, and leave the dishes under the biological
19 Compartmented Chambers for Studying Neurotrophic Factor Action 217
3. Methods
3.1. Setting Up the 1. Fill a 50-mL syringe with Corning vacuum grease. Use the
Compartmented syringe to fill the grease loader, wrap it in foil, and autoclave
Chambers (To Be for 45 min.
Done 1–2 Days Before 2. Make a scratch in the middle of a collagen-coated 35-mm ∅
Preparing Cells) dish with an outward motion. Scratching the substratum
removes the collagen and exposes the bare plastic beneath the
surface of the dish floor, which is a poor substratum for growth
cone attachment. This results in axon growth being confined
to the collagen-coated tracks formed between the scratches.
Axons are then directed to grow to the left and right where
they encounter the silicone grease barriers, grow under the
silicone grease, and emerge into the distal compartments (see
Note 3).
3. Place a drop of N2-methylcellulose solution on the middle of
the scratch. This prevents silicone grease from adhering directly
to the substratum in this zone when the Teflon divider is seated
(see Note 4). Set dish(es) aside until the divider is greased.
4. Attach a 23-gauge Luer Lock connector to the grease loader.
Grip the Teflon divider with a pair of 90° angle hemostatic
forceps and hold it horizontally with the divider facing up
under a binocular microscope. Outline the divider with grease.
Each time the adapter is placed at a new starting point, insert
the adapter into the grease from the previous step to assure a
continuous track of grease.
5. Once grease is applied to the entire divider, remove the lid
from one of the prepared 35-mm dishes, pick up the dish, and
quickly invert along the axis parallel with the scratches so that
the N2-methylcellulose droplet does not run.
6. While viewing the divider under the microscope, hold the dish
in position over the divider, oriented so that the scratches will
cross under the barriers between the proximal and distal com-
partments and placed so that the dish is resting gently on the
silicone grease Press down on the bottom of the dish with a
pair of tweezers. Make sure to press on the inside of the divider
at the four corners (see Note 5).
7. Using the pair of hemostatic forceps, turn the dish over and
release the divider (placing the dish on the work surface while
218 S.D. Skaper
3.2. Culturing of SCG The culture media formulations used are those described in the
or DRG Neurons relevant chapters of this volume, detailing protocols for preparing
rodent SCG or DRG cell cultures. The original formulation devel-
oped for rat SCG neurons uses rat serum (25), which is expensive
to obtain commercially and expensive and labor-intensive to prepare
in the laboratory. This formulation is not used here but, as pointed
out by Campenot and colleagues (10), some conditions may produce
diminished axon growth and may reduce the ability of axons to
cross into distal compartments. As recommended (10), methylcel-
lulose culture media are supplemented with methylcellulose, which
thickens the medium and reduces the shearing forces caused by
fluid movements that can perturb attachment of neurons to the
substratum. It facilitates wetting of the collagen before seating the
Teflon divider, and it prevents neurons from settling in the syringe
during plating.
1. Day 1: Replace culture medium in the side (distal) compartments
with medium containing 100 ng/mL NGF plus + Ara-C. Add
100,000 cells to center (proximal) compartment, using medium
containing 10 ng/mL NGF plus Ara-C (see Note 10).
2. Day 2: Add medium containing 10 ng/mL NGF plus Ara-C to
the outside of the Teflon divider until the medium flows over
the grease barrier and exchanges fluid with the center compart-
ment (see Note 11).
3. Day 3: Replace medium in the side compartments with medium
containing 100 ng/mL NGF but omitting Ara-C; replace
medium in the space outside of the Teflon divider with medium
containing 10 ng/mL NGF but omitting the Ara-C.
19 Compartmented Chambers for Studying Neurotrophic Factor Action 219
3.3. Analysis With experience, construction of the culture dishes should require
of Neurons about 3 h for 48 cultures, while preparing the neurons requires
3–4 h. Compartmented cultures from both SCG and DRG pro-
vide enough cellular material for biochemical analyses such as
immunoblotting, and cell bodies/proximal axons can be harvested
separately from distal axons. Morphological analyses have their
limitations, in that the presence of the Teflon divider produces
menisci in the surface of the culture medium that interfere with the
light path in inverted, phase-contrast microscopy. This can reduce
the contrast, making visualization of the neurons difficult, espe-
cially in the center compartment and close to the barriers in the
distal compartments. The menisci distort the ring of light produced
by the phase condenser so that it is not circular and therefore
cannot be aligned on the target annulus in the phase telescope.
Campenot and colleagues have solved this problem by constructing
a rotating shield that blocks ~80% of the ring of light, allowing
only a segment of the ring of light to pass through (10). Also, they
have modified the mechanical support of the light source/
condenser to increase the range of movement for adjustment in
the XY plane. These modifications make it possible to rotate the
shield and move the light source/condenser such that the segment
of the light ring that is not blocked by the shield projects nearly
completely onto the target annulus, which results in a reasonable
phase-contrast image. As the distortion arising from the meniscus
varies with location in the culture dish, adjustments must be made
whenever the field of view is moved. Also, the adjustments produce
better phase-contrast images at higher magnification, as the curva-
ture of the segment of the meniscus that must be compensated is
smaller, and therefore more uniform, when the field of view is
smaller. The need for this modification varies for different micro-
scopes. Tyler Research Corporation has provided Campenot and
colleagues with a modification for their Nikon Diaphot inverted
microscope. It is suggested that investigators intending to perform
such analyses liaise with Tyler for such modifications.
Recent studies have used microfluidic chambers instead of
these compartmented chambers. Microfluidic barriers are created,
which consist of narrow channels through which axons grow, such
that the extracellular space is kept to a minimum (26, 27).
Movement of substances across the barrier is controlled by estab-
lishing a flow through the extracellular space in the channels driven
by a pressure gradient produced by maintaining different fluid
220 S.D. Skaper
4. Notes
1. All dividers are machined from virgin Teflon and will last
for years if properly maintained. Many other types of dividers
are available or can be produced relatively quickly by the
company in response to an order. You can also provide a
drawing that they will cut to your specifications (biomedical@
tylerresearch.com).
2. Heat inactivation of fetal calf serum (and horse serum) is
recommended to destroy heat-labile complement. Thaw the
bottle of serum in advance, using a 37°C water bath. Next,
immerse the serum bottle in the water bath after re-equilibrating
to 56°C and leave for 30 min. Swirl the bottle occasionally to
ensure proper mixing. Allow the serum to cool to room
temperature, aliquot into 50-mL tubes, and store at −20°C.
3. The width of the collagen tracks is critical for axons to cross
into the distal compartments. The optimal track width for rat
sympathetic neuron cultures is about 200 μm (10). Extremely
narrow tracks are not conducive to axonal growth, and if tracks
are too wide, the growth cones have enough room to make
U-turns away from the barrier.
4. Do not let the medium used for wetting the collagen run
outside the area of the scratches before the divider is seated, as
this can interfere with sealing of the perimeter of the divider to
the dish. It is suggested to leave one or two tracks at each edge
dry, to reduce the risk of the droplet escaping beyond the
scratches (10). As the droplet does not extend the full distance
along the tracks, evaporation can leave a deposit on the sub-
stratum coinciding with the edge of the droplet across the
tracks, and which may interfere with axon growth. Construct
each culture dish and add medium covering the full length of
the tracks as soon as possible after placing the droplet.
5. It is important to press firmly enough so that the grease makes
a complete seal with the dish but if too much pressure is added,
the axons will not cross into the side compartments.
6. The dividers can be re-used after each experiment but must
first be cleaned. Remove the divider from the plate, wipe off all
19 Compartmented Chambers for Studying Neurotrophic Factor Action 221
References
1. Campenot RB (2009) NGF uptake and 15. Heerssen HM, Segal RA (2002) Location,
retrograde signaling mechanisms in sympa- location, location: a spatial view of neurotro-
thetic neurons in compartmented cultures. phin signal transduction. Trends Neurosci 25,
Results Probl Cell Differ 48, 141–158 160–165
2. Levi-Montalcini R (1987) The nerve growth 16. Watson FL, Heerssen HM, Bhattacharyya A,
factor 35 years later. Science 237, 1154–1162. Klesse L, Lin MZ, Segal RA (2001) Neurotrophins
3. Oppenheim, R.W. (1991) Cell death during use the Erk5 pathway to mediate a retrograde sur-
development of the nervous system. Annu Rev vival response. Nat Neurosci 10, 981–988
Neurosci 14, 453–501 17. Ng BK, Chen L, Mandemakers W, Cosgaya
4. Campenot RB (1977) Local control of neurite JM, Chan JR (2007) Anterograde transport
development by nerve growth factor. Proc Natl and secretion of brain-derived neurotrophic
Acad Sci USA 74, 4516–4519 factor along sensory axons promote Schwann
5. Ure DR, Campenot RB (1997) Retrograde cell myelination. J Neurosci 27, 7597–7603
transport and steady-state distribution of 18. Underhill SM, Goldberg MP (2007) Hypoxic
125I-nerve growth factor in rat sympathetic injury of isolated axons is independent of iono-
neurons in compartmented cultures. J Neurosci tropic glutamate receptors. Neurobiol Dis 25,
17, 1282–1290 284–290
6. Campenot RB (2009) NGF uptake and retro- 19. Hayashi H, Campenot RB, Vance DE, Vance
grade signaling mechanisms in sympathetic JE (2007) Apolipoprotein E-containing lipo-
neurons in compartmented cultures. in Results proteins protect neurons from apoptosis via a
and Problems in Cell Differentiation, Vol. 48 Cell signaling pathway involving low-density lipo-
Biology of the Axon (ed. Koenig, E.) pp 141–158 protein receptor-related protein-1. J Neurosci
(Springer, Berlin/Heidelberg) 27, 1933–1941
7. Zweifel LS, Kuruvilla R, Ginty DD (2005) 20. Sonderegger P, Fishman MC, Bokoum M,
Functions and mechanisms of retrograde Bauer HC, Neale EA, Nelson PG (1984) A few
neurotrophin signalling. Nat Rev Neurosci 6, axonal proteins distinguish ventral spinal cord
615–625 neurons from dorsal root ganglion neurons. J
8. Ginty DD, Segal RA (2002) Retrograde neu- Cell Biol 98, 364–368
rotrophin signaling: Trk-ing along the axon. 21. Eide FF, Lowenstein DH, Reichardt LF (1993)
Curr Opin Neurobiol 12, 268–274 Neurotrophins and their receptors–current
9. Campenot RB, MacInnis BL (2004) Retrograde concepts and implications for neurologic dis-
transport of neurotrophins: fact and function. J ease. Exp Neurol 121, 200–214
Neurobiol 58, 217–229 22. Ginty DD, Segal RA (2002) Retrograde neu-
10. Campenot RB, Lund K, Mok SA (2009) rotrophin signaling: Trk-ing along the axon.
Production of compartmented cultures of rat Curr Opin Neurobiol 12, 268–274
sympathetic neurons. Nat Protoc 4, 23. Campenot RB (1979) Independent control of
1869–1887 the local environment of somas and neurites.
11. Posse De Chaves EI, Vance DE, Campenot RB, Methods Enzymol 58, 302–307
Kiss RS, Vance JE (2000) Uptake of lipopro- 24. Heerssen HM, Pazyra MF, Segal RA (2004)
teins for axonal growth of sympathetic neurons. Dynein motors transport activated Trks to pro-
J Biol Chem 275, 19883–19890 mote survival of target-dependent neurons. Nat
12. Bertrand J, Winton MJ, Rodriguez-Hernandez Neurosci 7, 596–604
N, Campenot RB, McKerracher L (2005) 25. Mains RE, Patterson PH (1973) Primary cul-
Application of Rho antagonist to neuronal cell tures of dissociated sympathetic neurons. I.
bodies promotes neurite growth in compart- Establishment of long-term growth in culture
mented cultures and regeneration of retinal and studies of differentiated properties. J Cell
ganglion cell axons in the optic nerve of adult Biol 59, 329–345
rats. J Neurosci 25, 1113–1121 26. Taylor AM, Blurton-Jones M, Rhee SW, Cribbs
13. MacInnis BL, Campenot RB (2002) Retrograde DH, Cotman CW, Jeon NL (2005) A microflu-
support of neuronal survival without retro- idic culture platform for CNS axonal injury,
grade transport of nerve growth factor. regeneration and transport. Nat Methods 2,
Science 295, 1536–1539 599–605
14. Eng H, Lund K, Campenot RB (1999) 27. Park JW, Vahidi B, Taylor AM, Rhee SW,
Synthesis of beta-tubulin, actin, and other pro- Jeon NL (2006) Microfluidic culture platform
teins in axons of sympathetic neurons in com- for neuroscience research. Nat Protoc 4,
partmented cultures. J Neurosci 19, 1–9 2128–2136
Chapter 20
Abstract
Cultured chromaffin cells have been used for almost 40 years in the study of different cell functions using
biochemical, electrophysiological, pharmacological, and toxicological approaches. Chromaffin cells are
essentially secretory cells that are used to model sympathetic neurons or neuroendocrine cells. In this
chapter, we describe the most common methods currently used to isolate and culture chromaffin cells from
the animals used most commonly: cows, rats, and mice. We also provide some advice on the use of these
cells in the laboratory.
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_20, © Springer Science+Business Media, LLC 2012
223
224 N. Domínguez et al.
2. Materials
2.1. General Supplies, All procedures require standard culture room facilities with a
Solutions, and Culture laminar flow cabin, a Bunsen burner, a water bath set at 37°C
Medium (preferably with agitation), and a 37°C incubator with a water-
saturated atmosphere containing 5% CO2. An inverted microscope
with phase contrast and a low vibration clinical centrifuge are also
required. To plate chromaffin cells onto glass coverslips, we strongly
recommend high-quality glass Marienfeld (Lauda-Königshofen,
Germany). Other required materials and basic solutions include:
1. Sterile plastic Pasteur pipettes.
2. Syringes and 0.22-mm syringe filters.
3. Adjustable volume pipettes with sterile tips.
4. 15-mL conical centrifuge tubes (or 50-mL tubes for culturing
cow cells).
5. Hemocytometer (Neubauer).
6. 24-well culture plates.
7. Ca2+- and Mg2+-free Locke’s balanced salt solution: 154 mM
NaCl, 5 mM KCl, 3.6 mM NaHCO3, 5 mM HEPES, and
11 mM glucose.
8. Trypan blue: 4 mg/mL in Locke’s solution.
9. Culture medium: We always use standard Dulbecco’s modified
Eagle’s medium mixed 1:1 with Ham’s F12 and antibiotics to
culture chromaffin cells. The medium is completed by adding
10% fetal calf serum (Lonza, Basel, Switzerland, DE14-801 F,
see Note 1).
10. All the solutions used are sterilized by filtering through 0.22-
mm syringe filters and are supplemented to contain penicillin G
(100 IU/mL) and gentamicin sulfate (40 mg/L).
2.2. Materials 1. Sterile surgical material required: forceps, scissors, two scalpel
for Preparation blades, and one haemostatic clamp.
of Bovine Chromaffin 2. 2× 100 mm Ø Petri dishes.
Cells
3. Glass beakers: 2× » 30 mL, 1× » 100 mL, and 1× » 250 mL (for
waste).
4. Two »35-mm Ø glass funnels.
5. Standard cotton gauzes, »200-mm and »90-mm pore-size nylon
meshes.
6. 5-mL syringes and 0.22-mm sterilizing syringe filters.
7. For gradient purification, a refrigerated centrifuge capable of
reaching »8,000 × g is also required, along with two sterile
capped 30-mL transparent tubes suitable for centrifugation.
20 Preparation and Culture of Adrenal Chromaffin Cells 225
8. Collagenase IA.
9. Bovine serum albumin fraction V (BSA).
10. Deoxyribonuclease I (DNAase I).
11. Renografin® (ER Squibb & Sons, New Brunswick, NJ, USA)
or Urografin® (Schering España, Madrid, Spain) prepared at
15% in sterile pure water (this solution is isotonic).
3. Methods
3.1. Chromaffin Any of the methods described below for chromaffin cell isolation
Cell Preparation and culture are appropriate for amperometry, patch clamping, or
for Single-Cell fluorescent microscopy. The following is a rule of thumb for
Experiments amperometric studies using chromaffin cells of any species:
1. The isolation procedure should aim primarily to obtain healthy
cells rather than a high yield. We recommend reducing the
226 N. Domínguez et al.
3.2. Coverslips Glass coverslips can be sterilized by briefly flaming both sides using
and Adherent Support a Bunsen burner before placing them in culture plates. If it is nec-
Sterilization essary to coat them with a substrate (see Note 2), place a drop of
0.01% poly-D-lysine solution (Sigma, Catalog P-1024, prepared in
water) on each coverslip. Leave it for 20 min and then wash three
times with sterile pure water. Sterilize the coverslip by exposing it
to UV light in the fume hood for 30 min and use the treated glass
within 1 week of preparation.
3.3. Cell Viability Although many automatic systems for cell counting now exist, a
and Counting standard Neubauer hemocytometer is still a valuable and cheap tool
to count cells and to get a general idea of their viability. We prepare
a 1:9 dilution of cells by mixing 20 mL of cell suspension + 100 mL
of Locke’s solution + 80 mL of trypan blue staining solution (see
above). Guidelines for the correct use of hemocytometers have
been published elsewhere (7).
3.4. Procedure Glands may be obtained from the local abattoir. Choose only intact
for Bovine glands to avoid contamination and discard those with visible signs
Chromaffin Cells of internal blood coagulation. Do not remove the fat at the abattoir,
as it provides protection against contamination. The glands do not
require any special care for their transportation to the laboratory if
the time from sacrifice to culturing is less than 1 h. In the case of
longer intervals, it is advisable to inject 3–4 mL of Locke’s solution
into the glands and transport them in a plastic bag on ice. Other
protocols for bovine cell culture preparation have been published
elsewhere (4, 8–14).
Once in the culture room, wearing gloves spray the glands
with 60% ethanol under the laminar flow cabinet, making sure
that the solution does not enter the adrenal vein, and remove the
surrounding fat from the glands. The general procedure to mini-
mize contamination is to proceed progressively from a nonsterile
20 Preparation and Culture of Adrenal Chromaffin Cells 227
Fig. 1. Procedure to isolate and culture bovine chromaffin cells (see text for guidelines).
20 Preparation and Culture of Adrenal Chromaffin Cells 229
3.5. Procedure Animals older than »4 weeks produce a lower yield of cells than
for Adult Rat young rats. This is also observed with mice:
Chromaffin Cells
1. Prepare the dissociation solution (3–4 mL) and maintain it at
room temperature: collagenase type I (250–350 IU/mL),
3 mg/mL BSA, 0.15 mg/mL DNAase I, and 0.15 mg/mL
hyaluronidase I-S in Locke’s buffer.
2. Sacrifice the animals according to institutionally approved ethi-
cal procedures. Place the animal on its back and spray the
abdomen with 70% ethanol.
3. To access the abdominal cavity, perform an incision through
the abdominal wall, cutting the skin along the linea alba and
pulling it out to either side, using scissors to separate the skin
from underlying tissue. The abdominal cavity should tear open,
exposing the organs. Locate the two kidneys with the adrenal
glands on top, which should be readily visible (see Fig. 2a).
4. Remove the glands with fine-curved forceps and place them in
ice-cold Locke’s buffer.
5. Under a stereo dissection microscope coupled to a cold-light
source, remove the adipose tissue surrounding the gland with
a stainless steel scalpel blade and decapsulate the adrenal glands.
Next, remove the adrenal cortex to isolate the medullar tissue
and cut this into four pieces using a scalpel. Keep the tissue wet
throughout with cold Locke’s buffer.
20 Preparation and Culture of Adrenal Chromaffin Cells 231
Fig. 2. Procedure to isolate and culture mouse chromaffin cells (see text for guidelines).
3.6. Procedure We use animals of 2–4 weeks of age. Given the small amount of
for Young Mouse collagen in these adrenal tissues, papain is used instead of collagenase,
or Rat Chromaffin producing a considerably higher cell yield. As the quantity of starting
Cells material from the adrenal glands is very small, this procedure is
intended for single-cell measurements. We usually use 12-mm Ø
glass coverslips placed in 24-well plates, although other sizes can
be used. Figure 1 can be printed and posted in the culture room as
a brief guide to the procedure:
1. Prepare the papain solution in Locke’s buffer (12–18 IU in
200 mL, sufficient for four glands), and store it at 37°C.
2. Sacrifice the animal according to institutionally approved ethical
procedures, place it on its back, and spray the abdomen with
70% ethanol. Open the abdomen and remove both adrenal
glands by pulling the glands away with an angled forceps (see
Fig. 2a).
3. Under a stereo microscope (×20 magnification), carefully
remove the adrenal capsule as much as possible of the cortical
tissue (see Fig. 2b, c). Work on a sheet of clean filter paper
soaked with Locke’s solution. This helps to avoid drying and
overheating of the tissue by the light. Transfer the medullary
tissue to a sterile Petri dish (35 mm Ø) containing 1 mL of
sterile ice-cold Locke’s solution (see Fig. 2d).
4. Transfer the pieces to a 15-mL conical tube with 200 mL of
papain solution and leave them for 15–20 min at 37°C without
shaking. The incubation time should be adjusted according to
the age of the animals, as younger animals will require shorter
digestion.
5. Wash the tissue with 800 mL of fresh Locke’s solution.
6. Remove as much of the liquid as possible, taking particular
care as the tissue is almost digested, and then add 200 mL of
complete medium (see Note 1).
7. Disaggregate the tissue by triturating it several times, first
through a 1 mL pipette tip and then through a 100-mL pipette
20 Preparation and Culture of Adrenal Chromaffin Cells 233
4. Notes
Acknowledgments
References
1. Kloppenborg PW, Island DP, Liddle GW, culture of bovine chromaffin cells. Nat Protoc
Michelakis AM, and Nicholson WE (1968) A 2, 1248–1253
method of preparing adrenal cell suspensions 9. Krause W, Michael N, Lubke C, Livett BG, and
and its applicability to the in vitro study of adre- Oehme P (1996) Catecholamine release from
nal metabolism. Endocrinology 82, 1053–1058 fractionated chromaffin cells. Eur J Pharmacol
2. Hochman J, and Perlman RL (1976) Catecho- 302, 223–228
lamine secretion by isolated adrenal cells. 10. Livett BG, Boksa P, Dean DM, Mizobe F, and
Biochim Biophys Acta 421, 168–175 Lindenbaum MH (1983) Use of isolated chro-
3. Livett BG (1984) Adrenal medullary chromaffin maffin cells to study basic release mechanisms.
cells in vitro. Physiol Rev 64, 1103–1161 J Auton Nerv Syst 7, 59–86
4. Moro MA, Lopez MG, Gandia L, Michelena P, 11. Fenwick EM, Fajdiga PB, Howe NB, and Livett
and Garcia AG (1990) Separation and culture BG (1978) Functional and morphological char-
of living adrenaline- and noradrenaline- acterization of isolated bovine adrenal medullary
containing cells from bovine adrenal medullae. cells. J Cell Biol 76, 12–30
Anal Biochem 185, 243–248 12. Almazan G, Aunis D, Garcia AG, Montiel C,
5. Gilabert JA (2004) Necessary conditions to main- Nicolas GP, and Sanchez-Garcia P (1984)
tain rat adrenal chromaffin cells in primary culture. Effects of collagenase on the release of
In Cell Biology of the Chromaffin cell. Borges, R [3 H]-noradrenaline from bovine cultured
& Gandia, L. Eds. La Laguna. pp 269–274. web- adrenal chromaffin cells. Br J Pharmacol 81,
pages.ull.es/users/isccb12/TheBook.htm 599–610
6. Sorensen JB, Nagy G, Varoqueaux F, Nehring 13. Bader MF, Ciesielski-Treska J, Thierse D, Hesketh
RB, Brose N, Wilson MC, and Neher E (2003) JE, and Aunis D (1981) Immunocytochemical
Differential control of the releasable vesicle study of microtubules in chromaffin cells in cul-
pools by SNAP-25 splice variants and SNAP- ture and evidence that tubulin is not an integral
23. Cell 114, 75–86 protein of the chromaffin granule membrane.
7. Strober W (2001) Trypan blue exclusion test J Neurochem 37, 917–933
of cell viability. Curr Protoc Immunol Appendix 14. Baker PF, and Knight DE (1981) Calcium
3, Appendix 3B control of exocytosis and endocytosis in bovine
8. O’Connor DT, Mahata SK, Mahata M, Jiang adrenal medullary cells. Philos Trans R Soc Lond
Q, Hook VY, and Taupenot L (2007) Primary B Biol Sci 296, 83–103
Chapter 21
Abstract
Immunofluorescence is a technique allowing the visualization of a specific protein or antigen in cells or
tissue sections by binding a specific antibody chemically conjugated with a fluorescent dye such as fluorescein
isothiocyanate. There are two major types of immunofluorescence staining methods: (1) direct immunofluo-
rescence staining in which the primary antibody is labeled with fluorescence dye and (2) indirect immuno-
fluorescence staining in which a secondary antibody labeled with fluorochrome is used to recognize a
primary antibody. This chapter describes procedures for the application of indirect immunofluorescence
staining to neural cells in culture.
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_21, © Springer Science+Business Media, LLC 2012
235
236 M. Barbierato et al.
2. Materials
2.3. Solutions 1. Tris, 200 mM, pH 8.5: Dissolve 24.22 g of Tris base in 800 mL
of distilled deionized water in a beaker. Stir the mixture, and
adjust pH to 8.5 by adding concentrated HCl under a chemical
fume hood. Adjust the volume to 1 L by adding distilled deion-
ized water. Sterilize by autoclaving (see Note 1).
2. Phosphate buffer (PBS), 10×: 1.369 M NaCl (80 g/L), 26.827 mM
KCl (2 g/L), 100 mM Na2HPO4 (14.2 g/L, pH > 9.5), and
14.697 mM KH2PO4 (2 g/L). Add NaOH (1 N) to adjust pH
to 7.2 ± 0.2 at 25°C. Use diluted 10 times (1×).
3. 4% PFA in PBS: Warning—see Note 2. Dissolve 4 g of PFA in
50 mL of distilled deionized water in a beaker, under a chemi-
cal fume cupboard. Add 1 mL of 1 N of NaOH. On a stirring
hotplate, stir and heat (~65°C) the solution until the PFA is
completely dissolved, always under a chemical fume cupboard.
Add 10 mL of 10× PBS and allow the solution to cool at room
temperature. Check the pH (after cooling) using pH papers
(never the pH meter!), and, in case, adjust to pH 7.4 (at 25°C)
using 1 M HCl (~1 mL). Adjust the volume to 100 mL with
distilled deionized water. Filter the solution through a 0.45-
mm membrane filter (it is possible that some particles of the
power of PFA do not solubilize). The solution can be stored at
−20°C for several months. However, it is better to aliquot the
prepared stock solution. After use, do not freeze/thaw again.
Once thawed, the solution can be kept for up to 1 week at
+4°C. After this time, discard the aliquot.
4. Mowiol: Mowiol is a mounting medium. Weigh 6 g of Mowiol
4-88 and mix in a solution of 6 mL of glycerol and 6 mL of
distilled deionized water. Shake for 2 h at room temperature.
Add 12 mL of 200 mM Tris pH 8.5, and heat at 50°C for
about 3 h. Shake occasionally. Filter the solution through a
0.45-mm membrane filter, aliquot, and store at −20°C for 2
months (or at +4°C for 2 weeks).
5. “Antibody” buffer: 150 mM NaCl (2.25 g/200 mL), 50 mM
Tris base (1.5 g/200 mL), 1% (w/v) BSA (2 g/200 mL),
240 M. Barbierato et al.
2.4. Coating Glass 1. Heat 400 mL of distilled water to 60°C on stirring hotplate.
Microscope Slides 2. Dissolve 5 g of gelatin.
with Gelatin
3. Add 200 mg of chrome alum and cool.
4. Dip slides individually and allow to air dry by leaning them
vertically against a test tube rack.
5. Store in a slide box 4°C until used.
3. Methods
3.1. Coverslip Transfer 1. Using a coarse pair of forceps (keep cell side face up), transfer
to Staining Box coverslips from the 24-well plate to a staining box (a homemade
box with 24 flat-topped pedestals glued to its bottom to support
coverslips and keep them raised above the bottom which will
have some water for humidification). The box should also have
a top to prevent evaporation and keep slides humidified.
2. In the following steps, all volumes are 50–100 mL unless
otherwise specified. However, you can use as little as 25 mL if
necessary.
3.2. Fixation and 1. Place coverslips in the 4% PFA solution for 10 min at room
Permeabilization temperature, or 30 min at +4°C (except for bromodeoxyuri-
dine staining or other fixative-sensitive antigens, in which case
use 60–90 s maximum) (see Note 3). PFA cross-links proteins
so that they do not solubilize when the cells are permeabilized
(see below). Otherwise, fix in methanol or ethanol for 15 min
21 Indirect Immunofluorescence Staining of Cultured Neural Cells 241
3.4. Primary Antibody 1. Incubate the coverslip in the primary antibody solution for 2 h
Incubation at room temperature or overnight at 4°C (see Note 6). To
prepare this solution, dilute the primary antibody with 2%
serum in PBS or in “antibody” buffer. The best dilution
depends on the concentration of primary antibody and the
density of the antigen. Generally supernatants are used neat or
diluted up to 1:10. Ascites are used at 1:100 to 1:1,000, as are
polyclonal antisera. (The optimal dilution can be determined
in a separate experiment.) (see Note 7).
2. Rinse the coverslip five times in PBS.
3.5. Secondary 1. Incubate the coverslip in the secondary antibody solution for
Antibody Incubation 1–2 h. To prepare this solution, dilute the secondary antibody
with 2% serum in PBS or “antibody” buffer. The dilution
depends on the lot and concentration of the secondary anti-
body, the density of the antigen, and, to some extent, the con-
centration and the antigen affinity of the primary antibody that
was used.
2. A separate experiment can be used to establish the proper
antibody dilution. As a rule of thumb, the secondary antibodies
will generally be used at a final concentration of 20 mg/mL.
We suggest using a dilution of about 1:100 (1:500–1:1,000)
for secondary antibodies. For single-label staining, use fluorescein
242 M. Barbierato et al.
Fig. 2. Immunostaining of cortical astrocytes for GFAP. Astrocytes cultured from 2-day-old
neonatal rat cortex on poly-L-lysine-coated coverslips were fixed with paraformaldehyde.
Staining was performed using a mouse anti-rat GFAP monoclonal primary antibody, followed
by an Alexa Fluor® 555 goat anti-mouse secondary antibody. Images were captured
using a Leica DMI 4000B fluorescence microscope equipped with a Leica DFC480
camera, analyzed, and stored using Leica Application Suite software (version 2.8.1).
Magnification: ×400.
21 Indirect Immunofluorescence Staining of Cultured Neural Cells 245
4. Notes
References
1. Lakowicz JR (2006) Principles of fluorescence Rhodamine 6G by one- and two-photon
spectroscopy (3rd edition). Springer, New York, induced confocal fluorescence microscopy.
USA Chemphyschem. 6, 791–804
2. Monici M (2005) Cell and tissue autofluores- 6. Abbe E (1873) Archiv fuer Mikroskopische
cence research and diagnostic applications. Anatomie und Entwicklungs mechanik. 9,
Biotechnol Annu Rev 11, 227–256 413
3. Heikal AA (2010) Intracellular coenzymes as nat- 7. Liang R (2010) Optical design for biomedical
ural biomarkers for metabolic activities and mito- imaging. Spie Press, Washington USA
chondrial anomalies. Biomark Med 4, 241–263 8. Toomre D, and Bewersdorf J (2010) A new
4. Benson DM, Bryan J, Plant AL, Gotto AM Jr, wave of cellular imaging. Annu Rev Cell Dev
and Smith LC (1985) Digital imaging fluores- Biol 26, 285–314
cence microscopy: spatial heterogeneity of 9. Petibois C (2010) Imaging method for elemen-
photobleaching rate constant in individual tal, chemical, molecular, and morphological
cells. J Cell Biol 100, 1309–1323 analyses of single cells. Anal Bioanal Chem
5. Eggeling C, Volkmer A, and Seidel CA 397, 2051–2065
(2005) Molecular photobleaching kinetics of
Chapter 22
Abstract
High content analysis of neurite outgrowth enables the rapid and comprehensive phenotypic assessment
of individual neurons in a multiwell format amenable to high throughput assays. The resulting data are
considered “high content” because multiple measurements of neuronal outgrowth and morphometric
data are calculated from hundreds of individual cells within each image. This approach has been widely
adopted by the pharmaceutical industry to accelerate neurological drug discovery and in vitro safety assess-
ment. High content technology utilizes automated fluorescent and/or brightfield microscopy for image
acquisition. The acquired images are then quantified using mathematical algorithms to measure pertinent
neurobiological morphometric information, including neurite length, count, and extent of branching for
each cell within the images. Furthermore, evaluation of the individual cell-level measurements enables the
detection of subpopulations of cellular responders not apparent when examining well-level averages. Using
this technology, neurite outgrowth can be quantified in each well, derived from hundreds of cell measure-
ments in a 96-well microplate in approximately 30 min.
Key words: Neurite outgrowth, Immunofluorescence, High content analysis, High content screening,
Neuronal differentiation, In vitro neurotoxicity, PC12 cells
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_22, © Springer Science+Business Media, LLC 2012
247
248 N.M. Radio
Fig. 1. Early events in neurite outgrowth. (a) Diagram of early events in neurite outgrowth illustrating a cell body with lamel-
lipodia, the development of minor processes (tipped with a growth cone), and transformation of the processes into an axon
and dendrites. (b) Hoffman modulation contrast photomicrographs of neurite outgrowth in PC12 cells 2 h, 48 h, and 72 h
after treatment with 100 ng/mL NGF to induce differentiation. Reproduced with permission from Elsevier (5).
In comparison with the parent PC12 cell line, NS-1 cells are less
prone to cellular aggregation, allowing for the evaluation of neu-
rite outgrowth in individual cells, while retaining many of the
properties of PC12 cells (1). Previous studies using more tradi-
tional, manually derived methodologies demonstrate the ability of
a cell model to detect changes in neurite outgrowth; they were
mostly restricted to a single chemical analysis due to throughput
limitations (8, 9). For large volume chemical screening, fully auto-
mated techniques amendable to high-throughput analysis are nec-
essary to facilitate the rapid assessment of compound libraries.
Advances in automated microscopy combined with algorithm-
based quantification metrics have evolved into a cell-based mea-
surement approach called high content analysis. High content
analysis integrates quantitative microscopy with automated tech-
nology controlling image acquisition, quantified cell-level data
analysis, and data collection (10, 11). High content platforms are
designed to track phenotypic changes at the individual cell level in
multiwell plates using fluorescent labels and/or bright field imag-
ing. As a result of individual cell assessment, high content approaches
enable the detection of subtle heterogeneous effects not apparent
in average population responses (12). Furthermore, the increased
throughput abilities of high content combined with the microplate
formats enable the use of comprehensive dose-response curves to
characterize a concentration-dependent profile. For example, anal-
ysis in a 96-well plate enables a 12-point dose-response curve for
eight different compounds (see Fig. 2).
1 2 3 4 5 6 7 8 9 10 11 12
Fig. 2. Ninety-six-well plate chemical exposure format for neurite outgrowth evaluation. NS-1 cells are exposed to an
11-point dose-response curve (semilogarithmic concentration range from 1 nM to 100 μM) from eight separate chemicals
(Rows A–H ). Column 1 serves as a control column, with three replicates each of undifferentiated NS-1 cells (−NGF), cells
incubated with 100 ng/mL NGF (+NGF), and cells incubated with 100 ng/mL NGF coadministered with 3 μM bisindole-
maleamide-I (BIS-I) to serve as an internal pharmacologic control to evaluate expected neurite outgrowth inhibition. All
treatments contain 0.1% DMSO.
250 N.M. Radio
2. Materials
2.1. Cell Culture 1. NS-1 cells, a PC12 subclone (Thermo Fisher Scientific), are
Reagents maintained at 37°C in a 95% humidified incubator containing
5% CO2.
2. NS-1 cells are cultured in RPMI 1640 medium (Lonza
BioWhittaker, Walkersville, MD), supplemented with 10%
equine serum (HyClone, Logan, UT), 5% heat-inactivated fetal
bovine serum (HyClone), 1% L-glutamine (Lonza BioWhittaker),
1% penicillin/streptomycin (Lonza BioWhittaker), and 100 ng/
mL NGF (Sigma-Aldrich). After adding all components of the
medium, adjust the pH to 7.4 with HCl (see Note 1).
3. Human recombinant NGF (Sigma-Aldrich) is supplied as a
0.1 mg lyophilized powder. Dilute the powder with 500 μL of
0.1% bovine serum albumin in phosphate buffered saline
(PBS). Aliquot the 500 μL volume into 10-μL × 50-μL vials
and store at −80°C until time of use. On the day of use,
prepare NGF-supplemented medium (200 ng/mL) by adding
a 50 μL aliquot to 50 mL of RPMI medium.
22 Neurite Outgrowth Assessment Using High Content Analysis Methodology 251
2.3. Image Acquisition 1. Automated image acquisition is performed using the ArrayScan
and Analysis VTI high content imaging platform (Thermo Fisher Scientific).
2. Neurite outgrowth analysis is performed using the Neuronal
Profiling bioapplication (Thermo Fisher Scientific).
252 N.M. Radio
3. Methods
3.1. Cell Culture 1. A single passage number of NS-1 cells (i.e., ten after receipt
from supplier) should be used in every experiment to avoid
potential interpassage variability (15).
2. Grow NS-1 cells in T-75 flasks and trypsinize for use when
they reach 70–80% confluency (see Note 3).
3. To dislodge cells, aspirate the growth medium from the T-75
culture flask.
4. Add 3.0 mL of prewarmed (37°C) Trypsin-Versene solution to
the T-75 culture flask.
5. Gently tilt the flask so that the Trypsin-Versene solution covers
the cell surface. The first addition of Trypsin-Versene serves as
a wash step to remove the existing medium/serum and allows
for a smaller volume of Trypsin-Versene to be added.
6. After covering the cell surface of the flask with the 3.0 mL of
Trypsin-Versene, immediately aspirate the Trypsin-Versene
solution.
7. Add 1.0 mL of prewarmed (37°C) Trypsin-Versene solution to
the T-75 culture flask. Gently tilt the flask so that the Trypsin-
Versene solution covers the cell surface.
8. Incubate the cells in the Trypsin-Versene solution for 5 min at
37°C.
9. Add 9.0 mL of prewarmed complete medium directly to the
growth surface of the flask to dislodge the cells. Add the
medium to the growth surface of the flask several times to
ensure the cells are dislodged.
10. Transfer the cells into a 15-mL conical tube suitable for
centrifugation.
11. Centrifuge the cells at 300 × g for 5 min.
12. After centrifugation, remove the supernatant containing the
Trypsin-Versene solution and resuspend the pellet of cells in
3 mL of complete medium (see Note 4).
13. Perform a cell count using a hemocytometer to calculate the
cell density of the harvested cells.
14. Subdivide the harvested cells into two groups: −NGF-treated
cells (3 wells total) and +NGF-treated cells (93 wells total).
For each group, dilute the cells so the final density is 2,000
NS-1 cells per 90 μL. For the −NGF-treated cells, dilute the
cells in growth medium. For +NGF-treated cells, dilute the
cells in medium containing 100 ng/mL NGF.
15. Add 90 μL per well of the harvested cell suspension (containing
2,000 NS-1 cells) into a transparent, 96-well collagen IV–coated
22 Neurite Outgrowth Assessment Using High Content Analysis Methodology 253
3.3. Image Acquisition 1. The following instructions are based on use of the ArrayScan
VTI high content platform in conjunction with the iDEV™
Assay Development Workflow software in conjunction with
the Neuronal Profiling bioapplication. The method describes
automated measurement of neurite outgrowth in differenti-
ated cells in 96-well plates stained using the immunocytochem-
ical procedure described above. The assay parameters cited in
this method are associated with an optimized image analysis
protocol for measuring neurite outgrowth in differentiated
NS-1 cells. The notes associated with this method provide
guidance for adjustment of default settings and assay parameters
for adapting the protocol to measure neurite outgrowth in
other neuronal cell types (i.e., other cell lines, primary neu-
ronal cultures, etc.). Bold text refers to a menu or heading in
the Neuronal Profiling bioapplication. Italicized text refers to
an option or value input by the end-user (see Note 7).
2. Turn on the ArrayScan VTI HCS Reader, open the vHCS:Scan
Software, select the iDEV™ Assay Development Workflow mode,
and log in.
3. From the Select Protocol menu, click on the Change button
to select the NeuronalProfiling.V4 bioapplication.
4. Proceed to the Configure Acquisition parameters: Select the
×10 imaging Objective from the associated drop-down menu.
Set the Camera Configuration to an option that is compatible
with the ArrayScan system in use. Set Acquisition Camera
Mode to Standard. Set the number of channels to 2. The field
dimensions using these settings are 660.48 by 660.48 μm.
5. Specify the correct fluorophores to be utilized in the Image
Formation subbox: Use the drop-down menu to select XF100
– Hoechst as the Channel 1 dye and XF100 – FITC as the
Channel 2 dye.
6. In the XF100 – Hoechst Channel 1, select Fixed from the
Exposure Type drop-down menu. Set the Initial Exp Time
(s) to 0.01. In the XF100 – FITC Channel 2, select Fixed from
the Exposure Type drop-down menu. Set the Initial Exp
Time (s) to 0.03. These exposure values serve as initial expo-
sure settings that will used during initial image acquisition.
Acquired images deemed representative to the investigator
should be used to finalize exposure times, so nuclear images
22 Neurite Outgrowth Assessment Using High Content Analysis Methodology 255
use 25% of the camera’s dynamic range and the cell body/
neurite uses 55–65% (see Note 8).
7. Select Autofocus from the Autofocus Camera Mode drop-
down and specify Autofocus interval 3 fields (see Note 9).
8. Click on a representative undifferentiated (−NGF) control well
from the plate layout map. In cellular nuclei Channel 1, select
the Acquire Image button and then select Autofocus. Set the
final exposure value interactively by clicking the Autoexpose
button. Repeat this process for Channel 2 for cell body and
neurite image acquisition. Once suitable images are acquired
for both channels of the negative control well, select Save
Field so the image set is available for subsequent analysis.
9. Click a representative differentiated (+NGF) control well from
the plate layout map. Acquire images for both the nuclear
(Channel 1) and cell body/neurite (Channel 2) fluorophores.
Select Save Field so the image set is available for analysis. Select
Next to proceed to Configure Assay Parameters.
10. Within the Protocol Optimization Task List, proceed to the
Image Preprocessing step. Select the Object Type as Bright
for both Channels 1 and 2. Specify and enable the LowPassFilter
as the Background Removal Method for both Channels 1
and 2. The Value for both channels should be determined by
measuring the diameter of typically large valid objects (nucleus
in Channel 1 and cell body in Channel 2). The diameter of
these objects can be measured by using the status tray at the
bottom of the window. Due to a greater chance of cell body
size heterogeneity, double the diameter of the measured cell
body size.
11. Proceed to the Nucleus Identification Ch1 step. Enable
Smoothing, select the Uniform Method, and specify a
Value of 1.
12. For the Field Nucleus Identification Ch1, enable
Thresholding, select the Fixed Method, and determine a
Value by determining a difference (“delta”) between the
brightest background pixels and the dimmest foreground fluo-
rescence. Use half of this delta for a starting Value, and adjust
as needed, to produce accurate borders on the nucleus. Enable
Segmentation, selecting the Shape Method, and enter a
Value corresponding to half the diameter of a typical nucleus.
Object Cleanup should also be enabled to clean up the object
mask and remove small objects from identification.
13. Proceed to the Nucleus Validation Ch1 step. To be included
in analysis, any identified nucleus can be validated based on
size, shape, and fluorescence intensity measurements. Enable
the Nucleus.BorderObject.Ch1, Nucleus.Area.Ch1, and
Nucleus.ShapeLWR.Ch1 selection features. To exclude
256 N.M. Radio
Fig. 3. Differentiated NS-1 cells (100 ng/mL NGF, 96 h) image acquired with a ×10 objec-
tive lens and a ×0.63 coupler using the ArrayScan VTI. (a) Nuclei are stained blue with
Hoechst 33342 (5 ng/mL), and cell bodies and neurites are stained green using an anti-
βIII-tubulin primary antibody (1:800) and DyLight™ 488-congjugated antibody (1:500).
The resulting image was analyzed using the Neuronal Profiling bioapplication (b) for mor-
phometric data including cell body area, number of neurites, neurite length, and branch
point for each cell measured.
Fig. 4. Effects of neurite outgrowth inhibitors on NS-1 cells. NS-1 cells were plated at 2,000 cells per well in 100 ng/mL
NGF and evaluated for either total neurite length (closed circle) or viability (open square) following 96-h exposure to Bis-I.
Inset shows representative images of either control (0.1% DMSO) or 3 μM Bis-I-treated cells following the 96-h exposure.
Data are expressed as percent of the 0.1% DMSO control and are presented as means ± standard deviation from six total
wells analyzed across two independent experiments. Treatments that are Significantly different from control for total
neurite length (asterisk) or viability (multiplication sign) (one-way ANOVA followed by Dunnett’s test, p < 0.05). Reproduced
with permission from Oxford University Press (1).
4. Notes
Acknowledgments
References
1. Radio N, Breier J, Shafer T, Mundy W (2008) inhibition the site of action? Toxicol Appl
Assessment of chemical effects on neurite out- Pharmacol 160, 217–230
growth in PC12 cells using high content 9. Parran D, Mundy W, Barone S Jr (2001)
screening. Tox Sci 105, 106–118 Effects of methylmercury and mercuric chlo-
2. Craig A, Banker G (1994) Neuronal polarity. ride on differentiation and cell viability in PC12
Annu Rev Neurosci 17, 267–310 cells. Toxicol Sci 59, 278–290
3. Webb S, Monk C, Nelson C (2001) Mechanisms 10. Smith C, Eisenstein M (2005) Automated
of postnatal neurobiological development: imaging: Data as far as the eye can see. Nat
Implication for human development. Dev Methods 2, 547–555
Neuropsychol 19, 147–171 11. Dragunow M (2008) High-content analysis in
4. Ramakers G (2002) Rho proteins, mental neuroscience. Nat Rev Neurosci 9, 779–788
retardation and the cellular basis of cognition. 12. Giuliano K, Gough A, Taylor D, Vernetti L,
Trends Neurosci 25, 191–199 Johnston P (2010) Early safety assessment
5. Radio N, Mundy W (2008). Developmental using cellular systems biology yields insights
neurotoxicity testing in vitro: models for assess- into mechanisms of action. J Biomol Screen
ing neurite outgrowth. Neurotoxicology 29, 15, 783–797
361–376 13. Neely M, Nicholls J (1995) Electrical activity,
6. Vaundry D, Stork P, Lazarovici P, Eiden L growth cone motility and the cytoskeleton. J Exp
(2002) Signaling pathways for PC12 cell dif- Biol 198, 1433–1446
ferentiation: Making the right connections. 14. Harrill J, Robinette B, Mundy W (2011) Use
Science 296, 1648–1649 of high content image analysis to detect chemical-
7. Greene L (1977) A quantitative bioassay for induced changes in synaptogenesis in vitro.
nerve growth factor (NGF) activity employing Toxicol In Vitro 25, 368–387
a clonal pheochromocytoma cell line. Brain 15. Heumann R, Valet G, Maison D, Kemper J,
Res 133, 350–353 Reiser G, Hamprecht B (1977) Influence of
8. Das K, Barone S (1999) Neuronal differentia- the time in culture on cellular and neuronal
tion in PC12 cells is inhibited by chlorpyrifos properties of neuroblastoma x glioma hydrid
and its metabolites: Is acetylcholinesterase cells. J Cell Sci 27, 141–155
Chapter 23
Abstract
Cell culture has emerged as an important research method for studying the effects of carbon nanotubes
(CNTs) on primary neurons. We describe the procedure for preparation of dissociated mixed cell culture
from postnatal rat hippocampi. Based on morphological criteria and specific neuronal cell markers, neurons
can be selected within this mixed cell culture and studied. We present the procedure for the assessment
of neuronal cell morphology based on intracellular fluorescence of the vital dye calcein that accumulates
in live neurons. This procedure encompasses fluorescence imaging and measurement of the following
parameters: neurite number, total neurite length, mean neurite length, number of growth cones, number
of branches, and number of branches per neurite. These combined cell culture and fluorescence microscopy
approaches can be successfully used for assessment of the effects that CNTs, as water-soluble agents, have
on neuronal cell growth and neurite outgrowth.
Key words: Carbon nanotubes, Cell culture, Imaging, Morphology, Neurons, Growth, Neurite
outgrowth
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_23, © Springer Science+Business Media, LLC 2012
261
262 W. Lee and V. Parpura
2. Materials
2.3. Cell Culture 1. 0- to 2-day-old Sprague-Dawley rat pups (see Note 5).
2. Dissection tools (from Fine Science Tools): 10-mm angled
spring scissors, 2.5-mm angled spring scissors, Graefe curved
forceps (serrated), fine Dumont forceps (#55), and a blunt
curved glass probe (see Note 6). Dissection tools encased
within a Petri glass dish (Pyrex brand, 150 × 15 mm) (see Note 7)
need to be sterilized prior to use (autoclave; 121°C; 20 psi;
45 min).
23 Dissociated Cell Culture for Testing Effects of Carbon Nanotubes… 263
Fig. 1. Water-soluble CNTs used to investigate their effects on neuronal growth. Image of
two vials containing functionalized water-soluble SWNTs, SWNT-PEG (left ) and SWNT-
PABS (right ) at a stock concentration of 2 mg/mL, which appears as black liquid.
2.4. Cell Imaging 1. An imaging chamber with a circular recess at its bottom to
accommodate for mounting of a coverslip (see Note 11). Seal
the coverslip to the bottom of the chamber using Dow
Corning® High Vacuum Grease (see Note 12).
2. External solution: Prepare a saline solution containing 140 mM
NaCl, 5 mM KCl, 2 mM MgCl2, 2 mM CaCl2, and 10 mM
HEPES in water (pH = 7.4). Filter through a 0.2-μm filter and
store at 4°C. Immediately prior to use, warm it to room
temperature and add 5 mM D-Glucose (22.5 mg of glucose
per 25 mL of solution) to complete the external solution.
3. Calcein-AM stock solution: To make stock solution of calcein
acetoxymethyl (AM) ester (Invitrogen), add 50 μL of dry
dimethyl sulfoxide (DMSO) (see Note 13) to an individual vial
containing 50 μg of calcein-AM to yield a 1 μg/μL stock
solution. Store aliquots (1–10 μL) at −20°C.
4. Pluronic acid stock solution (25% w/v): Warm dry DMSO to
37°C. Weigh out 50 mg of pluronic acid F-127 (Invitrogen)
and add 200 μL of dry DMSO to it in a microcentrifuge tube
23 Dissociated Cell Culture for Testing Effects of Carbon Nanotubes… 265
3. Methods
3.2. Cell Culture The described procedures below, performed on rats, must be
approved by the Institutional Animal Care and Use Committee
prior to performing them. Hippocampal dissociated cell cultures
are prepared from 0- to 2-day-old Sprague-Dawley rats using pre-
viously described procedures; this cell culture approach yields a
mixed cellular population of neurons, astrocytes, and microglia,
and it has been used to study neuronal cell morphology (4–7, 9).
Since excellent photographs and video clips of rodent hippocampal
dissections are described elsewhere (10, 11), we only briefly and
textually address the isolation of hippocampi.
3.2.1. Preparatory Tasks 1. Prior to beginning a primary cell culture, place 50 μL of mito-MEM
onto each coverslip inlayed within 35-mm Petri dishes using
sterile technique in a laminar flow hood. Incubate dishes with
their lids on overnight inside the 95% air/5% CO2 atmosphere
incubator at 37°C. This step represents a convenient way to
allow the coverslip surface to wet properly and consequently
allow good cellular adhesion. If shorter lead time is desired, we
also have good results with cultures when medium was applied
onto PEI-coated coverslips ~4 h prior to application of dissoci-
ated cells; this is roughly ~2.5 h before the initiation of a the
dissection and culturing procedure (see Subheading 3.2.2).
2. Prepare the dissection area. Wear surgical gloves which should
be disinfected by spraying with rubbing alcohol (70% isopro-
panol) as needed. Take great care to clean the area (including
the dissecting scope, light source, and table top) thoroughly
with rubbing alcohol, which reduces the likelihood of contam-
ination. Place a terry cloth towel on the surface of the dissecting
area. This will protect the surface of the dissecting table from
debris; it will also prevent accidental sliding of dishes and
tools/beakers. Place two Kimwipe™ sheets at the bottom of
each of the two 200-mL beakers. This will protect tips of
dissections tools. Fill beakers with 70% isopropanol up to the
75 mL mark. In one beaker, place your gross dissecting tools
(10-mm angled spring scissors, Graefe curved serrated forceps,
and blunt curved glass probe), while in the other, place your
fine (Dumont forceps #55 and 2.5-mm angled spring scissors)
dissecting tools, with their tips (covered with pipette tips)
immersed in alcohol. Remove protecting pipette tips and shake
alcohol off any tool prior to its use on animal tissue.
3. Attend the media, solutions, and dishes. (a) Place a bottle of
fresh mito-MEM medium in an incubator to warm; this will
take ~3 h. Open the lid (unscrew ~½ of turn), but do not
remove it. This will allow pH equilibration. Medium can be
used for up to 2 weeks; however, the fresher the better. (b)
Also, remove a tube of papain solution (previously prepared in
1 mL aliquots) from a freezer and place in a hood to thaw.
23 Dissociated Cell Culture for Testing Effects of Carbon Nanotubes… 267
3.2.2. Dissection 1. Begin at a dissecting bench. Use sterile technique. Remove the
and Culturing pup from the transfer box, and spray its skin with alcohol.
Holding the pup’s front legs with your index finger and thumb,
position the pup with its back turned toward you. Using 10-mm
curved dissecting scissors, sever the spine/spinal cord at the
base of the skull to euthanize the pup. Then, make an incision
just under the skin and almost completely around the circum-
ference of the head. Peel the skin back to expose the skull.
Inserting the scissors through the hole created during the
euthanasia, cut the skull around the entire circumference of the
head. Be careful to keep the tips of the scissors pointed out so
as not to damage the brain beneath.
2. Lift the skull “cap” from the head using the Graefe forceps.
This should expose the brain. There may be a substantial amount
of blood bathing the brain at this point. Wash this blood
away using a few milliliters of HBSS from the 10-mL syringe.
While continuing to hold the pup, use the blunt probe to
dislodge the olfactory bulbs. This can be accomplished by
gently scooping (using the head of the hockey stick like glass
probe) the anterior end of the brain. Again using the blunt probe,
gently scoop the posterior end of the brain. This will separate
the brain from the spinal cord and allow it to be removed to a
culture dish containing 1 mL of cold HBSS.
3. Place the entire brain into a culture dish containing HBSS.
Separate the brain into two hemispheres by using the heel of a
fine dissecting scissors to “glide” from the midpoint of the cere-
bral cortex anteriorly through the front of the cortex between
the olfactory bulbs. With fine forceps, gently pry the hemispheres
apart to reveal the thalamus below. Again using the forceps,
pinch the remaining tissue between each hemisphere and the
thalamus to free them.
268 W. Lee and V. Parpura
15. Wash debris from each dish after the 3 h. Fill each dish with
1 mL of mito-MEM. Remove this mito-MEM with suction
and replace it with 1.0 mL of mito-MEM. For cultures that
examine the effect of water-soluble SWNTs, replace the dish
with 1.0 mL of mito-MEM with appropriate concentration of
SWNTs dissolved in the medium.
16. Maintain the cultures in an incubator (37°C; 95% air/5% CO2
atmosphere) for 3 days to allow for adequate growth, at which
point neuronal cell morphology can be assessed.
3.3. Cell Imaging 1. Inside a laminar flow biosafety hood, place a dish taken from
the 37°C incubator. Transfer one coverslip from a culture dish
3.3.1. Calcein Loading
into a 35-mm Petri dish containing loading solution, com-
posed of 1 μg/mL of calcein-AM and 0.025% of pluronic F-127
in external solution. Incubate the coverslip for 15 min at room
temperature (see Note 17). To transfer a coverslip, use a sterile
fine forceps with the tip flamed using an alcohol burner. Return
the Petri dish containing unused coverslips in culture media
back to the 37°C incubator immediately after transfer of an
experimental coverslip (see Note 18).
2. After the end of the 15-min incubation period in loading
solution, transfer the coverslip into another 35-mm Petri dish
containing 1 mL of external solution to allow for the dye to
de-esterify for 15 min.
3.3.2. Imaging 1. Prepare a clean imaging chamber for attachment of the cover-
and Analysis slip. After placing the chamber up-side down, apply a streak of
sealing grease at the recess of the chamber. Using sterile forceps,
take the coverslip containing cells out of external solution and
place it centered onto the chamber recess with the cell-side facing
toward the recess and lightly press it with forceps to loosely
adhere the coverslip to the grease. Flip the chamber so that the
open bath faces up, while the coverslip is at the bottom. Press
the chamber down against a Kimwipe™ to seal the coverslip
(see Note 19). Add ~400 μL of external solution (room tem-
perature) into the chamber. Check for leaks (see Notes 20 and
21). Aspirate the external solution and replace it with 400 μL
of external solution.
2. We use an inverted microscope (Nikon TE300) equipped with
differential interference contrast and epifluorescence illumina-
tion (xenon arc lamp, 100 W) to image the cells (see Note 22).
To visualize the calcein-loaded cells, we use a 60× plan apochro-
matic oil immersion objective (numerical aperture, 1.4; Nikon)
and a standard FITC filter set. We use a cooled charge-coupled
device camera (CoolSNAP HQ; Photometrics, Tucson, AZ)
270 W. Lee and V. Parpura
Fig. 2. Calcein-loaded neurons grown on PEI-coated glass coverslips and treated with chemically functionalized water-
soluble SWNTs added to the culture medium. Fluorescence images of live neurons, accumulating the vital stain, calcein.
Neurons grown on PEI-coated glass coverslips (control, sham treated) can be treated with CNTs (each at 1 μg/mL), either
SWNT-PABS (middle) or SWNT-PEG (right ) to affect their growth characteristics [consult the original work (7) for details].
Arrows indicate growth cones. Scale bar, 20 μm. Modified from ref. (7).
per neuron
4 2
* **
2 1
0 0
400 100
Total neurite length
** **
per neuron (µm)
300 75
200 50
100 25
0 0
60 15
Number of branches
Number of branches
*
per neuron
per neurite
40 10
20 5
0 0
control SWNT-PABS SWNT-PEG SWNT-PABS SWNT-PEG
0.1µg/mL 0.1µg/mL 1µg/mL 1µg/mL
(76) (60) (56) (44) (43)
Fig. 3. Parameters of neuronal growth and morphology. Neurons grown on PEI-coated glass coverslips (control) were
treated with SWNT-PABS or SWNT-PEG each at two different concentrations (0.1 and 1 μg/mL). Bars represent
means ± standard errors of means. Numbers in parentheses indicate the number of neurons studied in each condition.
Asterisks indicate a significant difference in measurements (*p < 0.05, **p < 0.01). For a description of the effects that
water-soluble SWNTs exert of neuronal morphology, consult the original work (7). Reproduced from ref. (7).
4. Notes
Fig. 4. Neurite tracing approach. Shown here are examples of neurite length measurements, where neurites were manually
traced by an investigator marking various loci of an individual neurite (ticks). Lengths of individual segments (between tick
marks) were summed to calculate the full length of a neurite (segmental lines).
Acknowledgments
References
Abstract
Dendritic spines act as sites of excitatory neuronal input in many types of neurons. Spine shape correlates
with the strength and maturity of synaptic contacts. Thus, evaluation of spine morphology is relevant for
studies on neuronal development, for determination of morphological correlates of learning and memory,
and for analysis of mechanisms of neurodegeneration. Here, we describe a method to determine spine
morphology in an ex vivo model of organotypic hippocampal slice cultures prepared from transgenic or
non-transgenic mice. Spines are imaged using confocal high-resolution imaging and evaluated by algorithm-
based analysis. The approach permits semiautomated determination of spine density and classification of
different spine types in dendritic segments from hippocampal subregions to evaluate intrahippocampal
connectivity.
Key words: Dendritic spine, Hippocampus, Organotypic culture, Laser scanning microscopy,
Automated morphology analysis
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_24, © Springer Science+Business Media, LLC 2012
277
278 F. Sündermann et al.
2. Materials
2.1. Components for 1. Animals: 6–8 days old mouse pups (e.g., mouse lines with
Slice Preparation, C57BL/6 background; see Note 1).
Culture, Infection, and 2. MEM: Minimum Essential Medium Eagle, HEPES
Fixation Modification without L-glutamine (Sigma-Adrich).
3. BME: Basal Medium Eagle with Earle’s, without L-glutamine
(Gibco BRL Life Technologies).
4. NB: Neurobasal Medium without L-glutamine (Gibco BRL
Life Technologies).
24 High-Resolution Imaging and Evaluation of Spines… 279
5. Horse serum.
6. Glutamine, 200 mM.
7. Pen-Strep: Penicillin/Streptomycin, 100× (PAA Laboratories).
8. Fungizone Gibco Fungizone® Antimycotic, liqid (Invitrogen).
9. N1-supplement: N1 medium supplement, 100×, for neural
cell cultures (Sigma-Adrich).
10. Virus: Virus constructs encoding fluorescent protein (e.g.,
SinRep5-eGFP) that can be optionally tagged with other pro-
tein of interest.
11. Confocal matrix : Micro-Tech-Lab (Graz, Austria).
2.2. Media, Prepare all media under sterile conditions using sterile filter such as
Supplements, 50 mL Steriflip or 500 mL Steritop-GP 0.22 μm ExpressTM
and Other Solutions Membrane (Millipore), depending on the volume of medium
needed.
1. Heat-inactivate horse serum at 56°C for 45 min. The aliquots
can be stored at −20°C for at least 1 year. (see Note 2).
2. Dissolve glucose powder in double distilled H2O (dd H2O) to
20% w/v and sterile-filter using a 0.22 μm filter. Store at 4°C.
3. Dissection medium: 1% glutamine and 1% Pen-Strep in MEM.
To 98 mL MEM, add 1 mL glutamine and 1 mL Pen-Strep.
The medium can be stored at 4°C for 24 h. (see Note 3).
4. Culture medium: 25% horse serum, 25% BME, 3% glucose, 1%
glutamine, 0.5% Pen-Strep, and 0.5% fungizone in MEM. In a
50 mL sterile conical tube add 12.5 mL horse serum, 12.5 mL
BME, 1.5 mL glucose, 0.5 mL glutamine, 0.25 mL Pen-Strep,
and 0.25 mL fungizone; bring volume to 50 mL with MEM
and filter sterilize. Preheat only the amount of medium that is
needed for a medium change on the same day (count 2–4 mL
additionally for pH adjustment). Adjust the pH value to 7.2
with HCl/NaOH. If not preheated, culture medium can be
stored at 4°C for at least 1 month. (see Note 4).
5. NB-N1 medium: 3% glucose, 1% glutamine, 1% N1-supplement,
0.5% horse serum, 0.5% Pen-Strep, and 0.5% fungizone in MEM.
In 50 mL sterile conical tube, add 1.5 mL glucose, 0.5 mL glu-
tamine, 0.5 mL N1-supplement, 0.25 mL horse serum, 0.25 mL
Pen-Strep, and 0.25 mL fungizone; bring volume to 50 mL with
NB and filter sterilize. Preheat only the amount of medium that
is needed for a medium change on the same day (count 2–4 mL
additionally for pH adjustment). Adjust the pH value to 7.2 with
HCl/NaOH. If not preheated, NB-N1 medium can be stored at
4°C for at least 1 month. (see Note 4).
6. Fixation solution: preheat 25 mL of phosphate buffed saline
(PBS) to 70°C. Add 1 g paraformaldehyde, mix and let it cool
down. Add 1 g sucrose. Store at −20°C.
280 F. Sündermann et al.
2.3. Culture and 1. Cell culture inserts. Millicell cell culture inserts, 30 mm, orga-
Dissection Dishes notypic hydrophilic PTFE, 0.4 μm. #PICM0RG50, Millipore.
(see Note 5).
2. Multidish 6-well plates (Nunclon™). Alternatively, single 3.5-cm
culture dishes can be used.
3. Bacterial dish (10 cm) as a dissection dish.
2.4. Dissection Tools 1. Large surgical scissors. Type: standard. Alloy: stainless steel.
(see Note 6; Fig. 1a) Tip shape: sharp/blunt. Tip angle: straight. Length: 13 cm.
Effective cutting edge: 42 mm. (#14001-13).
2. Small scissors. Type: standard. Alloy: stainless steel. Feature:
toughcut (this feature is not crucial). Tip shape: sharp/sharp.
(#14058-11).
3. Graefe forceps. Length: 10 cm. Alloy: stainless steel. Tip shape:
curved. Tips: serrated. Tip dimensions: 0.8 mm × 0.7 mm.
(#11052-10).
4. Dumont #5 Forceps. Length: 11 cm. Alloy: titanium (or inox).
Tip shape: straight. Tip dimensions: 0.05 mm × 0.02 mm.
(#11252-40).
5. Hippocampus tool (spatula) (×2). Length: 16 cm. Alloy: stain-
less steel. Tip shape: straight. Tip diameter: 8.5 mm × 0.3 mm.
(see Note 7).
6. Forceps. Length: 10.5 cm. Alloy: remanit 4301. Tip shape:
angled. Tips: serrated. (#2854.1).
7. Spatula (×2). Length: 13 cm. Alloy: stainless steel. End shape:
rounded. (see Note 7).
2.6. Equipment for Confocal Laser Scanning Microscope (e.g., Nikon Eclipse TE2000-U
Image Acquisition or Zeiss LSM 510) equipped with 10× and 20× air, 40× and 60×/63×
oil immersion objectives (suitable for fluorescence imaging), lasers
and filter sets corresponding to the fluorescent proteins (e.g., 488 nm
argon laser for GFP), and image acquisition software (e.g., EZ-C1
software, Nikon or LSM 5 software, Carl Zeiss).
Fig. 1. Preparation of organotypic hippocampal slices from mouse brain. (a) Preparation tools. (b) Dissection setup.
(c) Decapitation of a 1 week old mouse. (d) Incision of the skin along the midline of the head. (e) Removal of the skin to the
sides with the scissors. (f) Removal of the skull with the curved forceps. (g) Removal of the brain with the spatula.
(h) Cutting off the cerebellum. (i) Separation of two brain hemispheres with the spatula. (j) Dislodgement of the midbrain
with the spatula. (k) Hippocampus, prepared for cutting into slices. (l) Cutting of the hippocampus with the tissue chopper.
(m) Cut and separated slices. (n) Placing a slice onto a membrane insert within 6-well plate. (o) Three slices placed on the
membrane insert.
3. Methods
3.1. Preparation, Organotypic slice cultures from hippocampus are cultivated according
Culturing, Infection, to the membrane interface technique (4) (see Note 8).
and Fixation of
Hippocampal Slices
3.1.1. Dissection The procedure takes place under the sterile culture bench.
Preparation
1. Add 1 mL of culture medium to each well of a 6-well plate.
2. Place a membrane insert into each well with sterile forceps (see
Note 9).
3. Place the plates and dissection medium on ice.
4. Prepare two 50-mL centrifuge tubes with sterile PBS or ddH2O
(see Note 10).
5. Prepare sterile eppendorf cups on ice if tissue collection for
genotyping is required.
6. Cut rounds from Whatman paper, place one into each of the
dissection dishes and sterilize under UV (see Note 11).
3.1.2. Dissection The procedure takes place under the sterile preparation bench. The
of the Hippocampi tools are sterilized with 70% ethanol or by autoclaving. Solid cool-
ing pads can ensure low temperature during dissection procedure
(see Fig. 1b).
1. Decapitate a 6–8 day old mouse and place the head into the lid
of the sterile dissection dish (see Note 12; Fig. 1c).
2. Spray the head with 70% ethanol to avoid contamination of the
samples originating from the surface of the skin.
3. Fix the head at the eyes with the angled forceps, and using
small scissors, make an incision in the skin along the midline of
the head (see Fig. 1d).
4. Remove the skin on both sides with the scissors (see Note 13;
Fig. 1e).
5. Cut in the middle of the skull with the curved forceps
pulling it neatly below the skull from rostral to caudal end
(see Note 14).
6. Move away the sectioned skull with forceps (see Fig. 1f).
7. Remove the brain with spatula by gently reaching below (in
between the brain and skull (see Note 15)) and lifting it up
(see Fig. 1g).
8. Place the brain into the prechilled dissection medium.
9. Gently fix the brain at the position of the cerebral cortices with
one of the spatulas and remove the cerebellum with the other
(see Fig. 1h).
24 High-Resolution Imaging and Evaluation of Spines… 283
10. Further using the spatula, make a sagittal cut between the two
hemispheres to separate them (see Fig. 1i).
11. Flip one of the hemispheres onto the convex side and remove
the thalamus and the basal ganglia situated on top of the hip-
pocampus under a dissection microscope (see Fig. 1j).
12. Cut out the hippocampus from the underlying cortex (see
Fig. 1k).
13. With fine forceps clean away the vessels around the hippocam-
pus (see Note 16).
14. Repeat the procedure with the other hemisphere.
15. Place the two hippocampi in a small petri dish with prechilled
dissection medium on ice.
3.1.3. Preparation The procedure takes place under the sterile preparation bench.
of Hippocampal Slices
1. Using the spatulas, place two hippocampi on a teflon stage of
with Tissue Chopper
the tissue chopper.
2. Aspirate the excess medium with help of a sterile pipette (see
Note 17).
3. Cut rapidly 400 μm thick slices with the tissue chopper (see
Note 18; Fig. 1l).
4. Transfer the slices back into the small petri dish and place them
on ice (see Note 19).
5. Identify intact individual slices under the dissection microscope
and transfer them with the small spatulas onto the membrane
inserts (see Note 20; Fig. 1n).
6. Change the culture medium below the inserts to fresh, cooled
medium (see Note 21).
7. Place the 6-well plate with slices into the incubator at 37°C
with 5% CO2.
3.1.4. Maintenance and The procedure takes place under the sterile culture bench.
Infection of the Slices
1. Exchange the culture medium in the culture dishes every 2–3
days (see Note 22).
2. On day 11 post-preparation, change the culture medium to
NB medium containing N1 supplement (see Note 23).
3. On day 12 post-preparation, apply the virus with the droplet
method (see Note 24).
3.1.5. Fixation of Cultures are fixed at day 3 postinfection to ensure the highest
Hippocampal Slices expression of the fluorescent protein and best signal-to-noise ratio
for appropriate image processing.
1. Let the slices remain attached to the culture plate membrane
to preserve hippocampal structure and rinse with PBS within a
6-well plate (see Note 25).
284 F. Sündermann et al.
3.2. Microscopy of 1. Locate the effectively infected slices using 10× objective
Hippocampal Slices through eyepiece.
2. With 20× objective identify the regions of the hippocampus
and the cells that will be imaged.
3. With 40× objective identify the dendritic branches of individ-
ual pyramidal neurons on the apical or basal side, respectively.
4. Image CA1 and CA3 pyramidal neurons with voxel size of
0.08 × 0.08 × 0.25 μm in the x–y–z directions with 60×/63×
objective. Adjust the image size according to the length and
shape of the imaged dendritic fragment (see Note 28).
3.3.2. Analysis with 3DMA Autodeblur is run on a PC with “Windows XP” as an operating
system. The original “3DMA” software is created for operating
under “Unix/Linux” systems. To perform 3DMA based analysis, a
“Windows XP” PC for deconvolution and a “Unix/Linux” PC
(Ubuntu 6.06 LTS) for 3DMA analysis is required. For data trans-
fer, a “Share” folder, which can be accessed by both operating sys-
tems, is recommended (see Note 30).
24 High-Resolution Imaging and Evaluation of Spines… 285
Fig. 2. Deconvolution settings dialog window. In this dialog window deconvolution settings
can be adjusted as described in the protocol (refer to Subheading 3.3.1, steps 4–7). The
shown preselections for spine analysis are recommended by the authors.
Fig. 3. Results of the deconvolution. (a) The deconvolution algorithms enhance edges
within images and can produce artefacts like black borders around bright image com-
pounds. (b) Other artefacts are shadows of brighter structures imprinted in dark planes of
their neighborhood. The intensity of these imprints is slightly higher than the background.
(c) Projection of an appropriately deconvolved image stack. Scale bar, 2 μm.
01:> create_wdir
02:> cd raw
03:> 3dmaswitch
04:> aqh2mtif <file_without .tif>
05:> 3dmaswitch
06:> cd ../cases
07:> 3dmaGUI_lnx
Fig. 5. Screenshot of 3DMA menus. (a) Applications essential for spine analysis are listed: “Spine Detection Routines,” “Edit
Detected Spines,” and “Static Spine Analysis”. (b) After choosing “Spine Detection Routines” default options are recommended.
13. Select the options as shown in (see Fig. 6): uncheck <Use fiducial
polygon>, select <Simple Thresholding>. Enter a threshold
level in <Enter lower threshold>. As an alternative, choose
<Indicator Kriging> and enter a lower and an upper threshold
level (see Note 31; Fig. 7).
14. Click <Save Simple Settings> to save the entered settings. After
confirmation start your analysis by clicking <Run>.
15. After finishing the processing of the file start a <New Run>,
choose <Dendritic Spine> as application, and select the <Edit
Detected Spines> routine for manual correction.
16. In the menu which appears select the file to edit by double
clicking. Deselect falsely detected spines and dendrites by right
mouse click (see Fig. 8). Click <Save> to finish the editing
process (see Note 32).
288 F. Sündermann et al.
Fig. 7. Effects of thresholding. (a,c,e) Comparison of different threshold levels for spine detection to visualize the effects of
thresholding. (b,d,f) Projections of thresholded images with its detected spines. (b) Image with threshold set too low shows
a bias toward stubby spines. (d) Optimally thresholded image with no bias toward any spine type. (f) Images with threshold
set too high shows segmented dendritic backbone and a bias toward thin and mushroom spine types.
Fig. 8. Cropped dialog window for manual editing of detected spines. On the left side in the full dialog view, an overview
projection of the dendritic segment with colored spines is shown.
24 High-Resolution Imaging and Evaluation of Spines… 289
17. To complete your analysis and obtain the statistical data, start
again a <New Run>, select the <Dendritic Spine> application,
and choose the <Static Spine Analysis> routine.
18. In this menu load your data by clicking <Load Data> and set
the output filename. Alternatively, change the output folder as
well. The default options are “../spine/detect/analysis.txt.”
19. The analysis file can be read with every text or word editor (see
Note 33).
4. Notes
9. Take care that air bubbles do not remain under the membrane.
The membranes will become completely transparent when
wetted.
10. One of the tubes with sterile PBS/ddH2O may serve to clean
most of the blood and tissue from the tools before placing
them into 70% ethanol, the other to rinse the tools from etha-
nol before continuing to use them for further dissection.
11. Preparing the whole package of dishes at once, and packing
them back into the original sterile bag after sterilization together
with Whatman paper ensures a ready dissection dish for many
preparations. The Whatman paper provides a better contrast
and a non-slippery surface during the dissection procedure.
12. Cut off an ~3 mm portion of the tail if genotyping is required.
The tail should be placed immediately into a sterile eppendorf
cup kept on ice. Pay attention that the dish in which the decap-
itation is taking place is cleaned throughout with 70% ethanol
after each mouse to avoid false-positive PCR results due to
cross contamination of the genetic material.
13. The upper and lateral skull should be completely clean and
accessible for further preparation.
14. Care must be taken to avoid injury during this procedure.
Pulling the forceps slightly upward, while sliding it might help
avoid injury to the brain.
15. During this move the cranial nerves are cut so that the brain
can be flipped out.
16. The remaining connective tissue might block the separation of
the chopped slices.
17. Avoid injuring the slices but gently correct their position
with the pipette tip to align them perpendicular to the axes
of the blade.
18. Set the blade arm of the chopper in the position closest to the
hippocampi to minimize the cutting time. Add a drop of cold
dissection medium onto the slices immediately after chopping.
19. During this procedure gently pick up the slices with help of the
big spatulas and try to separate the slices without injuring them
by fine, round movements of the spatulas close to one another
(see Fig. 1m). Keep the slices on ice for 30 min before continu-
ing the procedure.
20. Up to 3 or 4 slices can be cultured on one insert (see Fig. 1o).
21. This procedure takes place under the sterile culture bench. Suck
all the medium from the dish with a sterile glass pasteur
pipette during each of the medium changes otherwise the new
medium will increase in volume due to the residual medium
resulting in leveling above the membrane and eventually to
24 High-Resolution Imaging and Evaluation of Spines… 291
Acknowledgements
References
1. Harris KM, and Kater SB (1994) Dendritic dynamics of dendritic spines in memory and
spines: cellular specializations imparting both cognition. Trends Neurosci 33, 121–129
stability and flexibility to synaptic function. 4. Stoppini L, Buchs PA, and Muller D (1991) A
Annu Rev Neurosci 17, 341–371 simple method for organotypic cultures of ner-
2. Tackenberg C, Ghori A, and Brandt R (2009) vous tissue. J Neurosci Methods 37, 173–182
Thin, stubby or mushroom: spine pathology in 5. Tackenberg C, and Brandt R (2009) Divergent
Alzheimer’s disease. Curr Alzheimer Res 6, pathways mediate spine alterations and cell
261–268 death induced by amyloid-beta, wild-type
3. Kasai H, Fukuda M, Watanabe S, Hayashi- tau, and R406W tau. J Neurosci 29,
Takagi A, and Noguchi J (2010) Structural 14439–14450
24 High-Resolution Imaging and Evaluation of Spines… 293
6. Ehrengruber MU, Lundstrom K, Schweitzer 10. Feng G, Mellor RH, Bernstein M, Keller-Peck C,
C, Heuss C, Schlesinger S, and Gahwiler BH Nguyen QT, Wallace M, Nerbonne JM, Lichtman
(1999) Recombinant Semliki Forest virus JW, and Sanes JR (2000) Imaging neuronal sub-
and Sindbis virus efficiently infect neurons in sets in transgenic mice expressing multiple spectral
hippocampal slice cultures. Proc Natl Acad Sci variants of GFP. Neuron 28, 41–51
USA 96, 7041–7046 11. Andersen P (1983) Basic mechanisms of peni-
7. Bakota L, and Brandt R (2009) Live cell imag- cillin-induced epileptiform discharges. Prog
ing in the study of neurodegeneration. Int Rev Clin Biol Res 124, 3–13
Cell Mol Biol 276, 49–103 12. Gähwiler BH (1988) Organotypic cultures of
8. Shahani N, Subramaniam S, Wolf T, Tackenberg neural tissue. Trends Neurosci 11, 484–489
C, and Brandt R (2006) Tau aggregation and 13. Gähwiler BH, Capogna M, Debanne D,
progressive neuronal degeneration in the McKinney RA, and Thompson SM (1997)
absence of changes in spine density and mor- Organotypic slice cultures: a technique has
phology after targeted expression of Alzheimer’s come of age. Trends Neurosci 20, 471–477
disease-relevant tau constructs in organotypic 14. Rodriguez A, Ehlenberger DB, Dickstein DL,
hippocampal slices. J. Neurosci 26, 6103–6114 Hof PR, and Wearne SL (2008) Automated
9. Ehrengruber MU, and Lundstrom K (2007 ) Three-Dimensional Detection and Shape Classifi-
Alphaviruses: Semliki Forest virus and Sindbis cation of Dendritic Spines from Fluorescence
virus vectors for gene transfer into neurons. Microscopy Images. PLoS ONE 3, e1997
Curr Protoc Neurosci Chapter 4:Unit 4.22
Chapter 25
Abstract
Fusion of fluorescent probes to axonally transported proteins represents an established approach that
enables live imaging of axonal transport. In this approach, in vivo examination of fluorescent particle
dynamics provides information about the length, directionality, and the velocity by which axonally trans-
ported proteins travel along axons. Analysis of these parameters provides information about the distribu-
tion of axonal proteins and their dynamics in and between different subcellular compartments. Establishing
the movement behavior of amyloid precursor protein within axons indicated that live imaging approaches
offer the opportunity to significantly enhance our understanding of the biology as well as pathology of
axonal transport. This chapter provides a fluorescence-based procedure for measuring axonal transport of
APP in cultured newborn mouse hippocampal neurons.
Key words: Amyloid precursor protein, Axonal transport, Fluorescent probes, In vivo fluorescent
imaging, Primary hippocampal cell culture
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_25, © Springer Science+Business Media, LLC 2012
295
296 T.L. Falzone and G.B. Stokin
2. Materials
2.1. Primary 1. Cold Hank’s balanced salt solution (HBSS, Gibco). Store at
Hippocampal Neuron 4°C (see Notes 1– 4).
Cultures 2. Hank’s buffer: to 500 mL HBSS, add 0.4 g of D-glucose
(Sigma-Aldrich), 0.834 g of 4-(2-hydroxyethyl)-1-pipera-
zineethanesulfonic acid (HEPES, Sigma-Aldrich), 5 mL of
100× penicillin-streptomycin (Invitrogen) and filter-sterilize
using 0.22-μm-diameter filter cartridge. Store at 4°C.
25 Imaging Amyloid Precursor Protein In Vivo: An Axonal Transport Assay 297
3. Methods
3.1. Harvesting Cells 1. Primary hippocampal cell cultures are harvested from postnatal
from Hippocampi day 1 mice (see Note 6). Using fine forceps under a dissecting
scope, cut the skin above calvarium by a midline anteroposte-
rior incision. Separate from the underlying bones of the calva-
rium both skin flaps generated by the incision. Incise the cranial
bones in the top midline portion and chip away to expose the
brain.
2. Detach the brain from the cranial nerves, remove from the
skull, and cut in half. Use a small spoon to remove the hip-
pocampi from the brain and meninges, and place into cold
HBSS (see Note 7).
3. Under a tissue culture hood, wash the excised hippocampi with
10 mL of HBSS at 4°C and then digest in 0.22-μ-filtered
papain disaggregation buffer for 20 min with moderate
shaking at 37°C.
4. After digestion, wash hippocampi twice with 10 mL of
complete DMEM (see Note 8), triturate 12 times with a 1-mL
micropipette in 2 mL of complete DMEM, and plate on top of
poly-D-lysine-coated coverslips at 60,000 cells per well in a
24-well dish (see Note 9).
5. After allowing 2 h for neurons to attach, replace the complete
DMEM with 500 μL of complete Neurobasal medium.
Maintain cell cultures at 37°C in 5% CO2 incubator.
Fig. 1. Axonal transport movie registered in primary hippocampal neurons transfected with
APP-YFP using an inverted epifluorescence microscope. 100-ms frame image montage of
a movie registered in a transfected axon showing dynamics of APP-YFP vesicles. White
arrows indicate stationary (1, 4, 7), anterograde (2, 3) or retrograde (5, 6) moving vesicles,
respectively. Time corresponds to frames extracted from a 30-s movie. Bar equals 10 μm.
300 T.L. Falzone and G.B. Stokin
a
Anterograde Retrograde
30 sec.
10 um
b 1 2 3 4 5 6 7
10 um
Fig. 2. Kymograph generated from movie showing APP-YFP axonal transport dynamics.
(a) Kymograph generated from movie in Fig. 1 showing the distance displacement of
anterograde, stationary, and retrograde APP-YFP vesicles along 30 s. (b) Schematic
representation of the above kymograph in which all APP-YFP vesicles were tracked and
plotted. Straight, dashed, and dotted black lines correspond to anterograde (2, 3), retrograde
(5, 6), and stationary (1, 4, 7) particles, respectively. Bar equals 10 μm.
b
50
Particle Proportion (%)
40
30
20
10
1.4 40
1.2 35
Run length (µm)
30
Speed (µm/sec)
1
25
0.8
20
0.6
15
0.4
10
0.2 5
0 0
Fig. 3. Axonal transport measurements obtained from kymograph analysis. (a) Excel file showing calculations extracted
from lines plotted on kymograph from Fig. 2b. Each line was separated by decreasing angle and corresponds to antero-
grade, stationary, and retrograde particles. Distance, time, and speed are calculated from the kymograph. (b) Results of
average particle proportion (%), speed (μm/s), and run length (μm) calculated from Excel file. Black, gray, and white bars
correspond to anterograde, stationary, and retrograde transport, respectively.
302 T.L. Falzone and G.B. Stokin
4. Notes
Acknowledgements
References
1. Hirokawa N, and Noda Y (2008) Intracellular study in cultured hippocampal neurons. Mol
transport and kinesin superfamily proteins, Biol Cell 11, 1213–1224
KIFs: structure, function, and dynamics. 11. Goldsbury C, Thies E, Konzack S, and
Physiol Rev 88, 1089–1118 Mandelkow EM (2007) Quantification of amy-
2. Kang J, Lemaire HG, Unterbeck A, Salbaum loid precursor protein and tau for the study
JM, Masters CL, Grzeschik KH, et al (1987) of axonal traffic pathways. J Neurosci 27,
The precursor of Alzheimer’s disease amyloid 3357–3363
A4 protein resembles a cell-surface receptor. 12. Stokin GB, Lillo C, Falzone TL, Brusch RG,
Nature 325, 733–736 Rockenstein E, Mount SL, et al (2005)
3. Bertram L, Lill CM, and Tanzi RE (2010) The Axonopathy and transport deficits early in the
genetics of Alzheimer disease: back to the pathogenesis of Alzheimer’s disease. Science
future. Neuron 68, 270–281 307, 1282–1288
4. Goedert M, and Spillantini MG (2006) A cen- 13. Stokin GB, Almenar-Queralt A, Gunawardena
tury of Alzheimer’s disease. Science 314, S, Rodrigues EM, Falzone T, Kim J, et al
777–781 (2008) Amyloid precursor protein-induced
5. Cras P, Kawai M, Lowery D, Gonzalez-DeWhitt axonopathies are independent of amyloid-beta
P, Greenberg B, and Perry G (1991) Senile peptides. Hum Mol Genet 17, 3474–3486
plaque neurites in Alzheimer disease accumu- 14. Falzone TL, Stokin GB, Lillo C, Rodrigues
late amyloid precursor protein. Proc Natl Acad EM, Westerman EL, Williams DS, et al (2009)
Sci USA 88, 7552–7556 Axonal stress kinase activation and tau misbe-
6. Stokin GB, and Goldstein LS (2006) Axonal havior induced by kinesin-1 transport defects.
transport and Alzheimer’s disease. Annu Rev J Neurosci 29, 5758–5767
Biochem 75, 607–627 15. Araki Y, Kawano T, Taru H, Saito Y, Wada S,
7. Stokin GB, and Goldstein LS (2006) Linking Miyamoto K, et al (2007) The novel cargo
molecular motors to Alzheimer’s disease. J Alcadein induces vesicle association of kine-
Physiol Paris 99, 193–200 sin-1 motor components and activates axonal
8. Joachim CL, Duffy LK, Morris JH, and Selkoe transport. EMBO J 26, 1475–1486
DJ (1988) Protein chemical and immunocy- 16. Eva R, Dassie E, Caswell PT, Dick G, ffrench-
tochemical studies of meningovascular beta- Constant C, Norman JC, et al (2010) Rab11
amyloid protein in Alzheimer’s disease and and its effector Rab coupling protein contrib-
normal aging. Brain Res 474, 100–111 ute to the trafficking of beta 1 integrins during
9. Koo EH, Sisodia SS, Archer DR, Martin LJ, axon growth in adult dorsal root ganglion neu-
Weidemann A, Beyreuther K, et al (1990) rons and PC12 cells. J Neurosci 30,
Precursor of amyloid protein in Alzheimer dis- 11654–11669
ease undergoes fast anterograde axonal transport. 17. Bilsland LG, Sahai E, Kelly G, Golding M,
Proc Natl Acad Sci USA 87, 1561–1565 Greensmith L, and Schiavo G (2010) Deficits
10. Kaether C, Skehel P, and Dotti CG (2000) in axonal transport precede ALS symptoms
Axonal membrane proteins are transported in in vivo. Proc Natl Acad Sci USA 107,
distinct carriers: a two-color video microscopy 20523–20528
Chapter 26
Abstract
Although primary neuronal cell cultures are a valuable source of in vitro insight for many neurobiologists,
all current gene expression technologies for these cells have significant drawbacks. Some of these limitations
of current gene expression protocols include toxicity, transient expression, a requirement for postnatal
neurons, and/or low efficiency. To date, many types of experiments were not possible because of these
limitations. Here, we outline a methodology by which primary cultured neurons can be transduced at any
age, after plating, with virtually no toxicity and continued gene expression for the lifetime of the culture.
This method involves the use of adeno-associated viral vectors, which have the potential to be highly useful
for either upregulation or downregulation of single or multiple genes, including neurotrophins, other
neuroprotective genes, and neurotoxins.
Key words: Viral vector, Gene delivery, Cultured neurons, Neurotrophins, Immunocytochemistry,
Neuroprotection assays, Glutamate toxicity
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_26, © Springer Science+Business Media, LLC 2012
305
306 S.Z. Malik et al.
2. Materials
2.1. Poly-L-Lysine 1. Many cultured cells, especially neurons, will not adhere to plastic
Coating and or glass without prior coating of the surface. Poly-L-lysine
Preincubation (Peptides International) has a positive charge which attracts
of Tissue Culture the negative surface charge of the neurons to the plate or well.
Plates with or Without Poly-L-lysine is made up in borate buffer.
Glass Coverslips 2. Borate buffer: add a stir bar to 500 mL of endotoxin-free water
(BioWhittaker/Lonza) and set on a stir plate in the biosafety
hood to stir. Add 2.38 g boric acid and 1.27 g borax to the
water and continue stirring for approximately 15 min or until
all the powder is dissolved. Resterilize the solution through a
0.2-mm filter and store in a labeled, sterile bottle at room
temperature for up to 6 months.
3. Poly-L-lysine solution: dispense 100 mL borate buffer into a
sterile 500-mL screw-cap bottle under the hood. Add one
bottle (100 mg) poly-L-lysine (molecular weight 70–150 kDa,
Peptides International, Louisville, KY) to the borate buffer and
swirl until all the poly-L-lysine has dissolved in the buffer, about
2 min.
4. Preincubation medium: combine 500 mL of Neurobasal
medium and 5 mL (one bottle) of B27 (with antioxidants)
supplement.
26 The Use of Specific AAV Serotypes to Stably Transduce Primary… 307
2.3. Construction 1. A plasmid containing the AAV inverted terminal repeats (ITRs)
of AAV Genomic and polyA sequence that has been propagated in recA- compe-
Plasmid tent cells and sequenced.
2. Additional plasmids containing the expression elements of
interest.
3. Standard molecular cloning reagents, including restriction
enzymes and buffers, ampicillin and/or kanamycin, LB broth
and plates, agarose gels, 37°C shaking incubator.
4. Competent bacterial cell lines such as SURE2 or STABL2 that
are recA-. These must be kept at −80°C and thawed on ice a
few minutes before transformation.
5. Primers that allow sequencing of the final plasmid, especially
the ITRs.
2.4. Packaging of AAV 1. Contact information for vector preparation experts (see
Subheading 3.5 for recommendations).
3. Methods
3.1. Preincubation 1. Observing sterile technique, use sterile forceps to add one
of Plates and/or coverslip per well of a 24-well plate or five coverslips to a
Coverslips 35-mm-diameter tissue culture plate (Nunc). Alternatively, use
a 24-well plate with no coverslips. The specific setup depends
on the type of experiment you wish to perform.
2. Set the plates in a single layer on an incubator tray and keep
inside the hood at room temperature.
3. For a 35-mm plate, add 1.5 mL of the poly-L-lysine solution
and make sure the coverslips, if present, are submerged and
arranged in a single layer on the bottom of the dish. Store at
room temperature for 24 h.
4. The next day, aspirate the poly-L-lysine solution and add
1.5 mL of BioWhittaker water to each dish, again being sure to
submerge the coverslips, and arrange in a single layer on the
26 The Use of Specific AAV Serotypes to Stably Transduce Primary… 309
3.3. Generation of AAV 1. AAV is a small (20 nm), single-stranded, icosahedral, nonrepli-
Vectors for Neuronal cating DNA virus composed of a ~4.7-kb genome comprised
Cell Transduction: of one ITR on each end of the genome and two open reading
Design Considerations frames which generate the multiple genes necessary for the
AAV life cycle and capsid production. Recombinant AAV vec-
tors are generated by packaging an engineered AAV genome
into an AAV capsid sequence that may or may not be derived
from the same AAV serotype as the genome. To generate
recombinant vectors, the rep/cap sequences can be delivered
in trans, allowing for more flexibility in vector design. Currently,
genome plasmids are generally based on the AAV2 ITRs
because they can be easily cross-packaged into capsids from
AAV1, 5, 6, 9, etc. Each capside sequence probably binds to a
separate receptor, though not all of them are known. These vec-
tors are called AAV2/5 or AAV2/9, etc., as specified by the
user. To generate the genome of the vector, the plasmid must
contain all the expression components in one continuous
sequence. (The total size of the entire plasmid is not critical
but must contain the antibiotic resistance gene, etc.) The usual
arrangement of AAV elements is 5¢ITR (usually derived from
AAV2) followed by a promoter, a gene of interest, a polyA
sequence, and the 3¢ITR. The entire sequence between the
5¢ITR and the 3¢ITR should not be much larger than the size
of the wild-type genome (~4.7 kb), otherwise the recombinant
genome will not be packaged efficiently. Unrelated “stuffer”
sequence is sometimes included if needed to bring the sequence
up to 4.7 kb. Maximal packaging size is about 5.2–5.3 kb (6–8).
To overcome some aspects of the size limitations, trans-splicing
vectors have been developed (for more details, see (9)).
26 The Use of Specific AAV Serotypes to Stably Transduce Primary… 311
3.4. Construction 1. To construct your genomic plasmid, begin with a plasmid con-
of AAV Genomic taining the ITRs and as many other components as desired.
Plasmid Many useful plasmids are available from academic investigators
and from an extensive AAV plasmid catalog available at the
National Gene Vector Biorepository (https://www.ngvbcc.
org/ReagentRepository.action), as well as from commercial
sources. Be sure a map is supplied and a sequence if possible. It
is highly advisable to sequence the AAV parts of the plasmid
before continuing.
2. It is ESSENTIAL to use competent bacteria that lack recA so
that the ITRs stay intact. For this, a literature search or pilot
experiment is necessary. If the provider of your source plasmid
has not done this, you can be sure that the ITRs are not com-
plete and you should not continue to use that plasmid.
Unfortunately, this is not an uncommon event. Again, this is
extremely important. Appropriate bacterial strains include
SURE2 and STABL2 cell lines which are recA-. Clone in your
promoter and cDNA of interest, as well as other elements. The
AAV genomic plasmids usually contain the polyA element
already; this should be confirmed. Again, you MUST use
competent cells lacking recA to propagate the intermediates.
Sequence the final product, especially the ITRs.
3. Confirm that the cassette is active by transfecting it into 293T
cells. Your gene of interest should be detectable by Western
blot, immunostaining, or other means.
3.5. Packaging of AAV 1. Methods for the packaging and purification of recombinant
AAV vectors are continually improving. For experiments using
neurons in vitro, clinical grade vectors are not required;
research grade vectors are sufficient. If financial resources are
available, we strongly recommend working with an experi-
enced core facility, such as the one at the University of
Pennsylvania (http://www.med.upenn.edu/gtp/vectorcore/),
the University of Michigan (http://www.med.umich.edu/
vcore/), or many other academic vector cores that package
AAV vectors for outside investigators. Alternatively, you can
contact a commercial AAV production company, although it
should be noted that packaging with certain serotypes is only
available at some companies. We would suggest that commer-
cial kits for packaging and purifying AAV vectors in your own
laboratory should be a last resort. We have observed significant
toxicity of purification by-products on neuronal cell cultures.
26 The Use of Specific AAV Serotypes to Stably Transduce Primary… 313
3.6.2. Addition 1. Two weeks after plating, add L-glutamate (0–400 mM) to the
of the Neurotoxic Agent wells, which should still contain the regular medium as well.
There should be a minimum of three replicates per experimen-
tal condition. As a control, transduce some cells but do not
treat with glutamate. These will be processed the same as the
other wells and will serve as the denominator for the calcula-
tion of percent toxicity (see below).
2. Twenty-four hours after addition of glutamate, aspirate the
medium and fix all cells with 10% neutral buffered formalin for
10 min at room temperature in a fume hood. Remove the for-
malin and dispose of it in chemical waste containers. Gently
wash the cells twice in PBS before proceeding. Add a small
drop of VECTASHIELD containing DAPI to each well. At
this stage, the plate can be sealed with parafilm and kept at 4°C
for up to a few days before imaging. Alternatively, the cover-
slips should be mounted on glass slides with the same mount-
ing medium and sealed with nail polish.
3.6.3. Quantification 1. An overall sense of the toxicity can be gained by a quick visual
of Neurotoxicity by Intrinsic inspection of the wells. We use a scale of 0–3, where 0 = healthy
GFP Fluorescence cultures with uniform GFP in the processes; 1 = beaded GFP
expression in the processes but little or no cell loss; 2 = substantial
cell loss and/or fragmented GFP fluorescence in the processes
and cell body; and 3 = majority of cells dead.
2. After that, live cells are quantified. Carefully seal the plate with
parafilm to prevent leakage of any fluid or mounting medium.
In an initial experiment, we obtained digital images of three non-
overlapping fields at 10× with the plate inverted on the micro-
scope stage, which was set to its lowest setting to accommodate
the plate (or with slides mounted normally on the microscope).
3. Manually count cells on each image, three nonoverlapping
fluorescent 10× fields per well and three wells per condition.
To insure reproducibility, it helps to have multiple lab mem-
bers count the cells. Calculate cell survival as percentage of the
untreated wells.
4. Immunostaining is also possible (see Subheading 3.7).
26 The Use of Specific AAV Serotypes to Stably Transduce Primary… 315
3.6.5. Determine 1. Plot the dose of glutamate against the percentage of live cells.
the Dose-Response Curve Choose the concentration of glutamate to use in all the follow-
ing neuroprotection assays. We prefer a concentration of gluta-
mate that represents about 30% cell survival so that we can
(1) determine whether a neuroprotective response is significant;
(2) determine whether there may be an unexpected result of
neurotoxicity by a supposedly neuroprotective construct; and
(3) have the potential to see significant intermediate degrees of
316 S.Z. Malik et al.
3.7. Quantification 1. This should be performed with a vector of the same serotype as
of Neuroprotection the reporter vector, but containing the gene of interest. This
technique does not require a GFP-containing vector (although
it does not preclude the use of one). Surviving cells are detected
by immunostaining for the protein of interest.
2. Fix the cells as described above but do not use mounting
medium.
3. A general immunostaining procedure is followed. After fixation,
permeabilize with TBST and block in 5% goat or donkey serum
in TBS. Expose to the primary antibody overnight at 4°C,
wash and incubate with the corresponding secondary antibody,
wash and incubate with the fluorescent streptavidin (1 h at
room temperature each), and then wash again and invert onto
glass slides with VECTASHIELD mounting medium contain-
ing DAPI to identify nuclei. If two antibodies are to be used at
the same time, note that one should be directly conjugated and
the other should be biotinylated. Similarly, the species of the
two antibodies should be different so that the secondary anti-
bodies do not bind to both primary antibodies. As to the fluo-
rescent tags, they should not be in the green range (488 nm) if
a GFP-expressing vector is being used and not in the blue
range (350 nm) to avoid conflict with the blue signal from the
DAPI. In general, we use a red fluorescent tag for the neurons
and, if necessary, a fluor in the 633-nm range for the second
antibody. After the last wash, apply VECTASHIELD mounting
medium with DAPI, about one drop per well. This mounting
medium does not set well, so it is essential to seal the plate well
or to invert the coverslips (if present) on a glass slide, dry, and
seal with nail polish.
4. For this protocol, neurons are identified by immunostaining
with rabbit polyclonal anti-microtubule-associated protein 2
(1:500) and detected with Biotin-SP-conjugated goat anti-
rabbit IgG and Alexa594-conjugated streptavidin (1:500 each).
Astrocytes are identified on separate coverslips with monoclo-
nal anti-GFAP antibody (1:500), Biotin-SP-conjugated donkey
anti-mouse IgG, and Alexa594-conjugated streptavidin (1:500
each). Confirm the specificity of staining by omitting the pri-
mary antibody in each case. Immunofluorescence is colocalized
26 The Use of Specific AAV Serotypes to Stably Transduce Primary… 317
4. Notes
Acknowledgments
References
1. Howard DB, Powers K, Wang Y and Harvey BK 8. Wu Z, Yang H and Colosi P (2010) Effect of
(2008) Tropism and toxicity of adeno-associated genome size on AAV vector packaging. Mol
viral vector serotypes 1, 2, 5, 6, 7, 8, and 9 in rat Ther 18:80–86
neurons and glia in vitro. Virology 372:24–34 9. Yan ZY, Zhang YL, Duan DS and Engelhardt
2. Kugler S, Lingor P, Scholl U, Zolotukhin S and JF (2000) Trans-splicing vectors expand the
Bahr M (2003) Differential transgene expres- utility of adeno-associated virus for gene ther-
sion in brain cells in vivo and in vitro from apy. Proc Natl Acad Sci USA 97:6716–6721
AAV-2 vectors with small transcriptional control 10. Shevtsova Z, Malik JM, Michel U, Bahr M and
units. Virology 311:89–95 Kugler S (2005) Promoters and serotypes:
3. Royo NC, Vandenberghe LH, Ma JY, Hauspurg targeting of adeno-associated virus vectors for
A, Yu L, Maronski M, Johnston J, Dichter MA, gene transfer in the rat central nervous system
Wilson JM and Watson DJ (2008) Specific AAV in vitro and in vivo. Exp Physiol 90:53–59
serotypes stably transduce primary hippocam- 11. Xu R, Janson CG, Mastakov M, Lawlor P,
pal and cortical cultures with high efficiency Young D, Mouravlev A, Fitzsimons H, Choi
and low toxicity. Brain Res 1190:15–22 KL, Ma H, Dragunow M, Leone P, Chen Q,
4. Goslin K, Asmussen H and Banker G 1998 Rat Dicker B and During MJ (2001) Quantitative
hippocampal neurons in low-density culture. comparison of expression with adeno-associ-
In: Culturing nerve cells. Banker G, Goslin K ated virus (AAV-2) brain-specific gene cassettes.
(eds). MIT Press: Cambridge, MA Gene Ther 8:1323–1332
5. Cummings DD, Wilcox KS and Dichter MA 12. McCarty DM, Monahan PE and Samulski RJ
(1996) Calcium-dependent paired-pulse facili- (2001) Self-complementary recombinant adeno-
tation of miniature EPSC frequency accompa- associated virus (scAAV) vectors promote
nies depression of EPSCs at hippocampal efficient transduction independently of DNA
synapses in culture. J Neurosci 16:5312–5323 synthesis. Gene Ther 8:1248–1254
6. Dong JY, Fan PD and Frizzell RA (1996) 13. Brun S, Faucon-Biguet N and Mallet J (2003)
Quantitative analysis of the packaging capacity Optimization of transgene expression at the
of recombinant adeno-associated virus. Hum posttranscriptional level in neural cells:
Gene Ther 7:2101–2112 implications for gene therapy. Mol Ther
7. Grieger JC and Samulski RJ (2005) Packaging 7:782–789
capacity of adeno-associated virus serotypes: 14. Michel U, Malik I, Ebert S, Bahr M and Kugler S
Impact of larger genomes on infectivity and (2005) Long-term in vivo and in vitro AAV-
postentry steps. J Virol 79:9933–9944 2-mediated RNA interference in rat retinal
26 The Use of Specific AAV Serotypes to Stably Transduce Primary… 319
ganglion cells and cultured primary neurons. 17. Cearley C and Wolfe JH (2006) Transduction
Biochem Biophys Res Commun 326:307–312 characteristics of adeno-associated virus vectors
15. Xia H, Mao Q, Eliason SL, Harper SQ, Martins expressing cap serotypes 7, 8, 9 and Rh10 in
IH, Orr HT, Paulson HL, Yang L, Kotin RM and the mouse brain. Mol Ther 13:528–537
Davidson BL (2004) RNAi suppresses polyglu- 18. Klein RL, Dayton RD, Tatom JB, Henderson
tamine-induced neurodegeneration in a model KM and Henning PP.(2008) AAV8, 9, Rh10,
of spinocerebellar ataxia. Nat Med 10:816–820 Rh43 vector gene transfer in the rat brain:
16. Boulaire J, Balani P and Wang S (2009) effects of serotype, promoter and purification
Transcriptional targeting to brain cells: Engi- method. Mol Ther 16:89–96
neering cell type-specific promoter containing 19. National Research Council (1996) Guide for the
cassettes for enhanced transgene expression. Care and Use and Use of Laboratory Animals.
Adv Drug Deliv Rev 61:589–602 National Academy Press: Washington, DC
Chapter 27
Abstract
Here, we describe a nanocarrier system that can transfer chitosan nanoparticles loaded with either small
peptides such as the caspase inhibitor Z-DEVD-FMK or a large peptide like basic fibroblast growth factor
across the blood–brain barrier. The nanoparticles are selectively directed to the brain and are not measurably
taken up by liver and spleen. Intravital fluorescent microscopy provides an opportunity to study the penetration
kinetics of nanoparticles loaded with fluorescent agents such as Nile red, and has demonstrated that this
nanomedicine formulation is rapidly transported across the blood–brain barrier.
Key words: Nanoparticles, Brain drug delivery, Blood–brain barrier, Neurotrophic factors, Caspase
inhibitors, Neuroprotection
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_27, © Springer Science+Business Media, LLC 2012
321
322 S. Caban et al.
Fig. 1. Schematic outline of nanoparticle preparation. Peptides (Z-DEVD-FMK or bFGF) or Nile red are loaded into CS–PEG–BIO
polymers and then nanosperes are formed by ionic gelation with tripolyphosphate solution (TPP). The nanoparticles are
conjugated with anti-mouse transferrin monoclonal antibodies (TFRMAb) by means of biotin–streptavidin bonds (reprinted
and modified from ref. (7) with permission).
27 Preparation and Characterization of Biocompatible Chitosan… 323
2. Materials
5. N-benzyloxycarbonyl-Asp(OMe)-Glu(OMe)-Val-Asp(OMe)-
fluoromethyl ketone (Z-DEVD-FMK) (BACHEM, USA):
Centrifuge a vial containing 1 mg of the peptide and then
dissolve the contents in 75 μL dimethyl sulfoxide. Divide the
solution into aliquots. Next, dilute each aliquot with water and
further divide into smaller working aliquots. Store at −20°C
until use (see Note 5).
6. Centrifuge a vial containing 50 μg of bFGF (Invitrogen, USA)
and then dissolve the contents in 500 μL water to obtain a
solution of 100 μg/mL. Divide this solution into smaller
working aliquots and store at −20°C (see Note 5).
7. Nile red: prepare a 1 mg/mL solution in dimethyl sulfoxide.
3. Methods
Fig. 2. Chemical structures of chitosan (Chi) and sodium tripolyphosphate, and schematic illustration of chitosan nanoparticle
preparation procedure with ionotropic gelation method. (TPP: tripolyphosphate solution). (Modified from ref. (28) with
permission).
3.2. Conjugation 1. Add 1,900 μL of borate buffer onto 100 μL of Traut’s reagent
of TfRMAb to and stir for 5 min on a magnetic stirrer.
Nanoparticles 2. Take equal volumes of the above and of the streptavidin
solution and mix together on a magnetic stirrer for 90 min to
obtain thiolated streptavidin.
3. In parallel, stir 100 μL of TfRMAb solution (1 mg/mL) with
5 μL of m-maleimidobenzoyl-N-hydroxysuccinimide solution
(5 mg/mL in dimethylformamide) for 30 min at room tem-
perature. This will transform some of the amino groups into
maleimide groups.
326 S. Caban et al.
Table 1
Examples of nanoparticle size and zeta potential
CS (blank) 301 22
CS (Z-DEVD-FMK loaded) 650 20
CS (bFGF loaded) 424 23
Table 2
Examples of drug association efficiency and loading
capacity of nanoparticles
Fig. 3. Scanning Electron Microscopy (SEM) images of loaded and PEGylated chitosan
nanoparticles.
27 Preparation and Characterization of Biocompatible Chitosan… 327
3.3. Release Studies 1. Add 1 mL of PBS in each release compartment after discarding
from Nanoparticles supernatant.
2. Place each eppendorf tube in a water bath at 37°C on a horizontal
shaker.
3. Centrifuge nanoparticles at 10,000 rpm (9,277 g) for 10 min
and then filter at predetermined time intervals (see Note 6).
4. Determine the concentration at each time point and calculate
the cumulative values (see Fig. 4).
3.4. In Vivo Monitoring 1. Anesthetize mice with isoflurane during surgery and with
of Nanoparticle urethane (750 mg/kg, intraperitoneal, followed by 500 mg/kg
Penetration 30 min later) during the experiment.
into the Brain 2. Monitor body temperature with a rectal probe and maintain at
37.0 ± 0.2°C using a homeothermic blanket control unit
(Harvard Apparatus). Monitor systolic blood pressure nonin-
vasively by using a cuff and tail probe (NIBP controller;
ADInstruments). Monitor pulse rate and oxygen saturation by
an oxymeter using amini Y clip on the left lower extremity
(V3304 Tabletop Pulse Oxymeter; Surgivet) (see Note 7).
3. Open a cranial window of 5 × 5 mm over the parietotemporal
cortex and leave the dura intact to maintain physiological con-
ditions. Seal the window with dental acryl and then fill it with
artificial cerebrospinal fluid (124 mM NaCl, 5 mM KCl, 1.25 mM
NaH2PO4, 1.3 mM MgSO4, 2.4 mM CaCl2, 25 mM NaHCO3,
and 10 mM glucose; pH 7.4) at 37°C. (see Notes 8 and 9).
328 S. Caban et al.
Fig. 5. Nanoparticles are rapidly transported to brain parenchyma after systemic administration. (a) The graph illustrates
the change in fluorescence recorded from the brain over the course of 3 h after injection of Nile red loaded to TfRMAb-
conjugated or unconjugated (TfRMAb-free) nanoparticles. The difference between the two lines (triangles) reflects the fluo-
rescence coming from the nanoparticles within the parenchyma and illustrates the time course of nanoparticle penetrance
to the brain. (b) Nile red concentration in brain postvascular tissue increased only when TfRMAb-conjugated nanoparticles
were administered. The graph illustrates spectrophotometric measurements at 549 nm from brain homogenates obtained
1 h after injection of TfRMAb-conjugated or unconjugated nanoparticles or from sham-operated mice. Only values above
the horizontal thick line, below which values correspond to the tissue background readings, were taken into consideration.
*p < 0.05 compared with the other groups. (c) Confirmation of penetration of the nanoparticles to the parenchyma by fluo-
rescence microscopy on brain sections obtained 1 h after injection. FITC-conjugated anti-rat IgG antibody (green) labeled
the nanoparticles bearing TfRMAb, clearly demonstrating that the nanoparticles were dispersed within the extracellular
space (arrowheads) outside the vessel lumens (longitudinal structures). Some FITC-conjugated nanoparticles exhibited
green as well as red fluorescence because they had not released of all the Nile red loaded within an hour (images on the
right ). Scale bars: (c), left, 15 μm; (c), right, 5 μm. (Reprinted from ref. (7) with permission).
27 Preparation and Characterization of Biocompatible Chitosan… 329
4. Notes
level over the window and the lighting conditions are constant
while recording sequential fluorescent images.
11. For histological detection of nanoparticles in the brain paren-
chyma, it is preferable to use two markers fluorescing at different
wavelengths (e.g. anti-IgG (green) and Nile red) and co-localize
both signals to ensure that the observed spots are not micro-
scopic staining artifacts.
Acknowledgements
References
1. Abbott, N. J., Ronnback, L., and Hansson, E. nanomedicine transports a peptide caspase-3
(2006) Astrocyte-endothelial interactions at inhibitor across the blood-brain barrier and
the blood-brain barrier, Nature Reviews provides neuroprotection, J Neurosci 29,
Neuroscience 7, 41–53. 13761–9.
2. Pardridge, W. M. (1998) CNS drug design 8. Chang, J., Jallouli, Y., Kroubi, M., Yuan, X. B.,
based on principles of blood-brain barrier Feng, W., Kang, C. S., Pu, P. Y., and Betbeder,
transport, J Neurochem 70, 1781–92. D. (2009) Characterization of endocytosis of
3. Bickel, U., Yoshikawa, T., and Pardridge, W. transferrin-coated PLGA nanoparticles by the
M. (2001) Delivery of peptides and proteins blood-brain barrier, Int J Pharm 379, 285–92.
through the blood-brain barrier, Adv Drug 9. Skarlatos, S., Yoshikawa, T., and Pardridge, W.
Deliv Rev 46, 247–79. M. (1995) Transport of [125I]transferrin
4. Pardridge, W. M. (2002) Drug and gene deliv- through the rat blood-brain barrier, Brain Res
ery to the brain: the vascular route, Neuron 36, 683, 164–71.
555–8. 10. Lee, H. J., Engelhardt, B., Lesley, J., Bickel,
5. Huwyler, J., and Pardridge, W. M. (1998) U., and Pardridge, W. M. (2000) Targeting rat
Examination of blood-brain barrier transferrin anti-mouse transferrin receptor monoclonal
receptor by confocal fluorescent microscopy of antibodies through blood-brain barrier in
unfixed isolated rat brain capillaries, J mouse, J Pharmacol Exp Ther 292, 1048–52.
Neurochem 70, 883–6.
11. Jefferies, W. A., Brandon, M. R., Hunt, S. V.,
6. Pardridge, W. M., Wu, D., and Sakane, T. Williams, A. F., Gatter, K. C., and Mason, D. Y.
(1998) Combined use of carboxyl-directed (1984) Transferrin receptor on endothelium of
protein pegylation and vector-mediated blood- brain capillaries, Nature 312, 162–3.
brain barrier drug delivery system optimizes
brain uptake of brain-derived neurotrophic fac- 12. Pardridge, W. M., Buciak, J. L., and Friden, P.
tor following intravenous administration, M. (1991) Selective transport of an anti-trans-
Pharm Res 15, 576–82. ferrin receptor antibody through the blood-
7. Karatas, H., Aktas, Y., Gursoy-Ozdemir, Y., brain barrier in vivo, J Pharmacol Exp Ther
Bodur, E., Yemisci, M., Caban, S., Vural, A., 259, 66–70.
Pinarbasli, O., Capan, Y., Fernandez-Megia, 13. Rawat, M., Singh, D., and Saraf, S. (2006)
E., Novoa-Carballal, R., Riguera, R., Andrieux, Nanocarriers: promising vehicle for bioactive
K., Couvreur, P., and Dalkara, T. (2009) A drugs, Biol Pharm Bull 29, 1790–8.
332 S. Caban et al.
14. Peracchia, M. T., Fattal, E., Desmaele, D., stroke and neurodegenerative diseases, Annals
Besnard, M., Noel, J. P., Gomis, J. M., Appel, of Neurology 45, 421–29.
M., d’Angelo, J., and Couvreur, P. (1999). 22. Chen, J., Nagayama, T., Jin, K. L., Stetler, R.
Stealth PEGylated polycyanoacrylate nanopar- A., Zhu, R. L., Graham, S. H., and Simon, R.
ticles for intravenous administration and splenic P. (1998) Induction of caspase-3-like protease
targeting. J Control Release 60, 121–8. may mediate delayed neuronal death in the hip-
15. Prabaharan, M. (2008) Review paper: chitosan pocampus after transient cerebral ischemia,
derivatives as promising materials for controlled Journal of Neuroscience 18, 4914–28.
drug delivery, J Biomater Appl 23, 5–36. 23. Thornberry, N. A., and Lazebnik, Y. (1998)
16. Fernandez-Urrusuno, R., Calvo, P., Remunan- Caspases: Enemies within, Science 281, 1312–16.
Lopez, C., Vila-Jato, J. L., and Alonso, M. J.
24. Aktas, Y., Yemisci, M., Andrieux, K., Gursoy,
(1999) Enhancement of nasal absorption of
R. N., Alonso, M. J., Fernandez-Megia, E.,
insulin using chitosan nanoparticles, Pharm
Novoa-Carballal, R., Quinoa, E., Riguera, R.,
Res 16, 1576–81.
Sargon, M. F., Celik, H. H., Demir, A. S.,
17. Pan, Y., Li, Y. J., Zhao, H. Y., Zheng, J. M., Hincal, A. A., Dalkara, T., Capan, Y., and
Xu, H., Wei, G., Hao, J. S., and Cui, F. D. Couvreur, P. (2005) Development and brain
(2002) Bioadhesive polysaccharide in protein delivery of chitosan-PEG nanoparticles func-
delivery system: chitosan nanoparticles improve tionalized with the monoclonal antibody
the intestinal absorption of insulin in vivo, Int J OX26, Bioconjugate Chemistry 16,
Pharm 249, 139–47. 1503–11.
18. Vila, A., Sanchez, A., Tobio, M., Calvo, P., and
25. Illum, L. (1998) Chitosan and its use as a phar-
Alonso, M. J. (2002) Design of biodegradable
maceutical excipient, Pharm Res 15, 1326–31.
particles for protein delivery, J Control Release
78, 15–24. 26. Caban, S., Yemisci, M., Gursoy-Ozdemir, Y.,
19. Datta, S. R., Brunet, A., and Greenberg, M. E. Fernandez-Megia, E., Novoa-Carballal, R.,
(1999) Cellular survival: a play in three Akts, Riguera, R., Andrieux, K., Couvreur, P., Capan,
Genes Dev 13, 2905–27. Y., and Dalkara, T. (2010) in “AAPS Annual
Meeting and Exposition”, New Orleans,
20. Hara, H., Friedlander, R. M., Gagliardini, V., Louisiana, USA.
Ayata, C., Fink, K., Huang, Z. H.,
ShimizuSasamata, M., Yuan, J. Y., and 27. Ma, J., Qiu, J., Hirt, L., Dalkara, T., and
Moskowitz, M. A. (1997) Inhibition of inter- Moskowitz, M. A. (2001) Synergistic protec-
leukin 1 beta converting enzyme family pro- tive effect of caspase inhibitors and bFGF
teases reduces ischemic and excitotoxic against brain injury induced by transient focal
neuronal damage, Proceedings of the National ischaemia, Br J Pharmacol 133, 345–50.
Academy of Sciences of the United States of 28. Cho, Y., Shi, R., and Ben Borgens, R. (2010)
America 94, 2007–12. Chitosan nanoparticle-based neuronal mem-
21. Schulz, J. B., Weller, M., and Moskowitz, M. brane sealing and neuroprotection following
A. (1999) Caspases as treatment targets in acrolein-induced cell injury, J Biol Eng 4, 2.
Chapter 28
Abstract
The concentration of nerve growth factor (NGF) is elevated in a number of inflammatory and autoimmune
states in conjunction with increased accumulation of mast cells. Mast cells, which are of hematopoietic
lineage, and NGF appear to be involved in neuroimmune interactions and tissue inflammation. Mast cells
themselves are capable of producing and responding to NGF. Here we describe a protocol for the isolation
and culture of peritoneal-derived rat mast cells, together with a [3H]serotonin release assay which is useful
in assessing the effects of antigens and neurotrophic factors on mast-cell activation.
Key words: Mast cells, Antigen, Degranulation, Serotonin, Nerve growth factor, Nerve–immune
interaction
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_28, © Springer Science+Business Media, LLC 2012
333
334 S.D. Skaper and L. Facci
2. Materials
2.2. Cell Culture Media, 1. Bovine serum albumin (BSA), Cohen fraction V (Sigma)
Supplements, and [ 3H] 2. RPMI-1640 medium (Invitrogen)
Serotonin Release
3. L-Glutamine (200 mM, sterile, for cell culture) (Invitrogen)
Assay Reagents
4. Penicillin/streptomycin (10,000 U/mL penicillin and
10,000 mg/mL streptomycin) (Invitrogen)
5. Trypan blue stain 0.4% (Invitrogen)
28 [3H]Serotonin Release Assay Using Antigen-Stimulated Rat Peritoneal Mast Cells 335
2.3. Culture Medium 1. PBS + 0.6 % (w/v) glucose: Dissolve 0.6 g of D-glucose in
and Solutions 100 mL of PBS and filter-sterilize.
2. 40 % (w/v) BSA in PBS/0.6 % glucose: Dissolve 40 g of BSA in
100 mL of PBS and filter-sterilize (see Note 1).
3. 30 % (w/v) BSA: Dilute the 40% glucose solution (75 mL) in
PBS/0.6% glucose (25 mL).
4. Culture medium: To 100 mL of RPMI-1640 medium, add
1 mL of 200 mM L-glutamine stock solution (2 mM final) and
0.5 mL penicillin/streptomycin 100× stock solution (50 U/
mL penicillin and 50 mg/mL streptomycin, final).
5. PIPES buffer pH 7.10: 25 mM PIPES (7.56 g/L), 100 mM
NaCl (5.84 g/L), 5 mM KCl (0.372 g/L), 1 mM CaCl2·2H2O
(0.147 g/L), MgCl2×6H2O (0.81 g/L), and 5.6 mM D-glucose
(1.00 g/L).
3. Methods
3.1. Isolation 1. Adult male CD Sprague Dawley rats (200–250 g) are used. The
of Mast Cells recommended procedure for euthanasia is CO2 asphyxiation
and cervical dislocation. This should be carried out in strict
compliance with approved institutional and national guidelines.
2. Using a 50-mL syringe (1.5-in. needle), inject into the perito-
neal cavity 40 mL of sterile PBS/0.6% (w/v) glucose.
3. Gently massage the abdomen for about 90 s.
4. Carefully open the peritoneal cavity and collect the fluid contain-
ing peritoneal cells. Take care to avoid entering internal organs,
which can contaminate the wash solution (see Notes 2 and 3).
336 S.D. Skaper and L. Facci
3.2. Safranin Mast cells can be divided into two types: connective tissue mast
O Staining cells and mucosal mast cells. Peritoneal mast cells belong to the
of Mast Cells first type. Mast-cell phenotypes can be classified in terms of their
profiles of proteinases and proteoglycans. In this regard, mucosal
mast-cell granules stain with Alcian blue rather than with Safranin,
while connective tissue mast cells stain with Safranin but not with
Alcian blue:
1. This procedure makes use of the Shandon Cytospin three
centrifuge (Block Scientific, Bohemia, NY, USA, www.
blockscientificstore.com).
28 [3H]Serotonin Release Assay Using Antigen-Stimulated Rat Peritoneal Mast Cells 337
3.3. Measuring [ 3H] Mast cells have high-affinity IgE receptors (FceRI) (19) and have
Serotonin Release been used extensively to study the signaling pathways leading to
from Antigen- exocytotic release of inflammatory mediators during antigen
Stimulated Mast Cells stimulation of mast cells (20, 21). In this assay, a mouse monoclo-
nal IgE which is specific for dinitrophenol (DNP) haptens (clone
SPE-7) is used. DNP-HSA is employed as the triggering agent in
this assay. Cells are preloaded with [3H]serotonin (5-[1,2-3H(N)]
hydroxytryptamine binoxalate) which is released upon FceRI
cross-linking.
1. Isolate rat peritoneal mast cells, as described in Subheading 3.1
above.
2. Plate mast cells in a 96-well tissue culture plate (brand is not
important) at a density of 50,000 cells per well in RPMI-1640
medium with 2 mM L-glutamine and 50 U/mL penicil-
lin + 50 mg/mL streptomycin + 0.1 mCi of [3H]serotonin. Final
volume: 100 mL per well.
3. Incubate cells for 18 h in a 5% CO2 incubator (37°C).
4. Replace this medium with 100 mL of PIPES buffer containing
anti-DNP IgE (0.3 mg/mL) to sensitize the cells.
5. Incubate for 1 h at 37°C.
6. Replace the IgE solution with 100 mL per well of prewarmed
PIPES buffer containing 0.1–1.0 mg/mL of DNP-HSA.
338 S.D. Skaper and L. Facci
3.4. Assay Application NGF is known to induce mediator release from mast cells (22).
to Study of NGF Effects of NGF on stimulus-evoked mediator release from mast
Regulation of Mast- cells can be assessed using the [3H]serotonin release assay described
Cell Function here. A representative experiment is now described, in which rat
peritoneal mast cells are incubated with NGF, together with spe-
cific antigen (15) or substance P (23) to induce degranulation. The
steps below are taken from Subheading 3.3 above:
1. Seed mast cells into a 96-well tissue culture plate, 50,000 cells
per milliliter.
2. Load with [3H]serotonin for 18 h.
3. Remove this solution and prime cells with anti-DNP IgE for
1 h at 37°C.
4. Replace the IgE solution with 100 mL of prewarmed PIPES
buffer containing DNP-HSA (0.1 mg/mL) or substance P
(30 mM).
5. In replicate wells containing DNP-HSA or substance P, add
the following neurotrophins to a final concentration of 100 ng/
mL: NGF, BDNF, NT-3, or NT-4. The neurotrophin is added
to the DNP-HSA or substance P–containing solution from a
25-mg/mL stock solution (i.e., 1:250) (see Note 6).
6. Continue incubation for a further 15 min.
7. Determine net release of [3H]serotonin.
8. Note that a significant increase in secretion of preloaded [3H]
serotonin occurred in mast cells treated with specific antigen or
substance P, in combination with NGF, but not BDNF, NT-3,
or NT-4 (see Table 1).
28 [3H]Serotonin Release Assay Using Antigen-Stimulated Rat Peritoneal Mast Cells 339
Table 1
Exogenous NGF regulates mast-cell function
4. Notes
Acknowledgments
References
1. Levi-Montalcini R (1987) The nerve growth 6. Otten U, Ehrhard P, and Peck R (1989) Nerve
factor 35 years later. Science 237, 1154–1162 growth factor induces growth and differentia-
2. Galli SJ, Nakae S, and Tsai M (2005) Mast cells tion of human B lymphocytes. Proc Natl Acad
in the development of adaptive immune Sci USA 86, 10059–10063
responses. Nat Immunol 6, 135–142 7. Matsuda H, Coughlin MD, Bienenstock J, and
3. Böhm A, Aloe L, and Levi-Montalcini R (1986) Denburg JA (1988) Nerve growth factor pro-
Nerve growth factor enhances precocious dif- motes human hemopoietic colony growth and
ferentiation and numerical increase in mast cells differentiation. Proc Natl Acad Sci USA 85,
in cultures of rat splenocytes. Accademia 6508–6512
Nazionale Lincei 80, 1–6 8. Torcia M, Bracci-Laudiero L, Lucibello M,
4. Aloe L, and Levi-Montalcini R (1977) Mast cells Rubartelli A, Cozzolino F, Aloe L, et al (1996)
increase in tissues of neonatal rats injected with Nerve growth factor is an autocrine factor for
nerve growth factor. Brain Res 133, 358–366 memory B lymphocytes. Cell 85, 1–20
5. Thorpe LW, and Perez-Polo JR (1987) The 9. Ehrhard PB, Ganter U, Bauer J, and Otten U.
influence of nerve growth factor on the in vitro (1993) Expression of functional trk protoonco-
proliferative response of rat spleen lymphocytes. gene in human monocytes. Proc Natl Acad Sci
J Neurosci Res 18, 134–139 USA 90, 5423–5427
28 [3H]Serotonin Release Assay Using Antigen-Stimulated Rat Peritoneal Mast Cells 341
10. Ehrhard PB, Erb B, Graumann U, and Otten U active high molecular weight neurotrophins.
(1993) Expression of nerve growth factor and Mol Brain Res 97, 177–185
nerve growth factor receptor tyrosine kinase 17. Skaper SD (2001) Nerve growth factor: a neu-
Trk in activated CD4-positive T-cell clones. rokine orchestrating neuroimmune-endocrine
Proc Natl Acad Sci USA 90, 10984–10988 functions. Mol Neurobiol 24, 183–199
11. Lambiase A, Bracci-Laudiero L, Bonini S, 18. Mayrhofer G (1980) Fixation and staining of
Bonini S, Starace G, D’Elios MM, et al (1997) granules in mucosal mast cells and intraepithe-
Human CD4+ T cell clones produce and release lial lymphocytes in the rat jejunum, with special
nerve growth factor and express high-affinity reference to the relationship between the acid
nerve growth factor receptors. J Allergy Clin glycosaminoglycans in the two cell types.
Immunol 100, 408–414 Histochem J 12, 513–526
12. Santambrogio L, Benedetti M, Chao MV, 19. Metzger H (1992) The receptor with high
Muzaffar R, Kulig K, Gabellini N, et al (1994) affinity for IgE. Immunol Rev 125, 37–48
Nerve growth factor production by lympho- 20. Benhamou M, Stephan V, Robbins KC, and
cytes. J Immunol 153, 4488–4495 Siraganian RP (1992) High-affinity IgE recep-
13. Horigome K, Pryor ED, Bullock EM, and tor-mediated stimulation of rat basophilic leu-
Johnson EM Jr (1993) Mediator release from kemia (RBL-2H3) cells induces early and late
mast cells by nerve growth factor. Neurotrophin protein-tyrosine phosphorylations. J Biol Chem
specificity and receptor mediation. J Biol Chem 267, 7310–7314
268, 14881–14887 21. Beaven MA, and Metzger H (1993) Signal
14. Horigome K, Bullock ED, and Johnson, EM Jr transduction by Fc receptors: the Fc epsilon RI
(1994) Effects of nerve growth factor on rat case. Immunol Today 14, 222–226
peritoneal mast cells. Survival promotion and 22. Bruni A, Bigon E, Boarato E, Mietto L, Leon
immediate-early gene induction. J Biol Chem A, and Toffano G (1982) Interaction between
269, 2695–2702 nerve growth factor and lysophosphatidylserine
15. Leon A, Buriani A, Dal Toso R, Fabris M, on rat peritoneal mast cells, FEBS Lett 138,
Romanello S, Aloe L, et al (1993) Mast cells 190–192
synthesize, store, and release nerve growth fac- 23. Fewtrell CM, Foreman JC, Jordan CC, Oehme
tor. Proc Natl Acad Sci USA 91, 3739–3743 P, Renner H, and Stewart JM (1982) The effect
16. Skaper SD, Pollock M, and Facci L (2001) ofsubstancePonhistamineand5-hydroxytryptam-
Mast cells differentially express and release ine release in the rat, J Physiol 330, 393–411
Chapter 29
Abstract
Organotypic slices cultured for weeks in vitro represent an extremely valuable strategy for the investigation
of the long-term properties of neuronal circuits under physiological and pathological conditions. Here, we
describe how to prepare rat organotypic hippocampal slice cultures and how to expose them for appropri-
ate periods of time to excitotoxic agents or to oxygen and glucose deprivation conditions, in order to
mimic the pattern of pyramidal cell damage which is observed in vivo and in other in vitro models. This
preparation is very useful not only to study synaptic plasticity or the pathways and mechanisms of neuro-
degeneration but also to evaluate the effects of neuroprotective agents.
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_29, © Springer Science+Business Media, LLC 2012
343
344 E. Gerace et al.
2. Materials
2.1. Dissecting This is the medium used for hippocampal dissection and slice prep-
Medium aration. The solution is prepared under a sterile laminar flow
hood:
heat gently to dissolve. Add water to 100 mL, filter (0.2 μm),
and collect into a sterile recipient. Aliquot in Eppendorf sterile
tubes and store at −20°C for 1–2 months.
2. Prepare approximately 100 mL of Dissecting Medium in two
50-mL Falcon sterile tubes placed in a test-tube rack under the
hood at room temperature: Add 50 mL of Hanks’ Balanced
Salt Solution (HBSS, Sigma H9269) (see Note 1) to each of
the Falcon tubes using a sterile pipette. Then add 0.5 mL of a
45% glucose solution to each of the tubes and 0.75 mL of
amphotericin B (see Note 2).
3. Vortex to mix all ingredients and store at 4°C for maximum
1–2 weeks.
2.2. Normal Medium This is the medium used to grow and maintain the hippocampal
slice cultures. Prepare approximately 100 mL of Normal Medium
in two 50-mL Falcon sterile tubes placed in a test-tube rack under
the hood at room temperature, as follows:
1. Add 25 mL of Eagle’s Minimal Essential Medium (MEM,
Sigma M2279) (see Note 3) to each of the Falcon tubes using
a sterile pipette. Then add 12.5 mL of HBSS to each of the
tubes, 12.5 mL of Horse Serum (see Note 4), 0.5 mL of a 45%
glucose solution, and 0.75 mL of amphotericin B.
2. Finally, add 0.25 mL of 200 mM glutamine (see Note 5) to
each of the tubes.
3. Vortex to mix all ingredients and store at 4°C for maximum
1–2 weeks.
4. Warm up to 37°C in a water bath before use.
2.3. Serum-Free This is the medium used for excitotoxicity experiments and for
Medium the 24-h recovery period following OGD and excitotoxicity exper-
iments. Prepare approximately 100 mL of Serum-Free Medium in
two 50-mL Falcon sterile tubes placed in a test-tube rack under the
hood at room temperature, as follows:
1. Add 37.5 mL of Eagle’s MEM to each of the Falcon tubes
using a sterile pipette. Then add 12.5 mL of HBSS to each of
the tubes, 0.5 mL of a 45% glucose solution, and 0.75 mL of
amphotericin B.
2. Finally, add 0.25 mL of 200 mM glutamine to each of the
tubes.
3. Vortex to mix all ingredients and store at 4°C for maximum
1–2 weeks.
4. Warm up to 37°C in a water bath before use.
346 E. Gerace et al.
2.5. Experimental 1. Propidium iodide (PI, Sigma P4170, see Note 6): Prepare
Reagents approximately 10 mL of a 1-mg/mL solution in sterile water,
filter (0.2 μm), aliquot in foil-wrapped Eppendorf tubes, and
store in a closed box at 4°C. PI is a highly polar compound
which is normally excluded from cells with an intact plasma
membrane. When the membrane is damaged, PI enters the
cells and upon binding to exposed DNA becomes highly fluo-
rescent (the absorption maximum is 535 nm, and the fluores-
cence emission maximum is 617 nm).
2. Glutamate (Sigma G8415): Prepare a 100-mM stock solution
in water. Add a few drops of 0.1 N NaOH to dissolve. Filter
(0.2 μm), aliquot in Eppendorf tubes, and store at −20°C.
3. N-methyl-D-aspartate (NMDA, Sigma M3262): Prepare a
100-mM stock solution in water. Filter (0.2 μm), aliquot in
Eppendorf tubes, and store at −20°C.
4. (S)-α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
(AMPA, Ascent Scientific 005): Prepare a 10-mM stock solu-
tion in water. Filter (0.2 μm), aliquot in Eppendorf tubes, and
store at −20°C.
5. Kainic acid (Tocris Bioscience 0222): Prepare a 20-mM stock
solution in water. Filter (0.2 μm), aliquot in Eppendorf tubes,
and store at −20°C.
2.6. Special Equipment 1. Laminar flow hood. Both a vertical or horizontal laminar flow
hood can be used.
2. Cell culture CO2 incubator. The conditions are 37°C and 100%
humidity in 95% air/5% CO2.
3. McIlwain tissue chopper (Mickle Laboratory Engineering Co.
Ltd., UK). Use a new double-edge blade (Gillette Platinum or
from the chopper manufacturer) for every slice preparation
session.
29 Rat Hippocampal Slice Culture Models… 347
3. Methods
3.1. Preparation 1. Under a sterile laminar flow hood, fill each well of 3 or more
of Hippocampal Slice 6-well sterile Corning cell culture plates (see Note 7) with
Cultures 1.2 mL of prewarmed (37°C) sterile Normal Medium. Place
one Millicell-®CM insert into each well and keep the plates in
an incubator at 37°C and 100% humidity in 95% air/5% CO2
until use.
2. Place the following items on an aluminum foil sheet under the
hood: one Whatman (Grade No. 1) filter paper disc (diameter:
9–10 cm) for each pup, one polyethylene disc (diameter: 2 cm)
for each pup (see Note 8), a dissecting scalpel with a new blade,
two pairs of scissors (one large and one small), two pairs of
small tweezers (one straight and one curved), 3–4 paddle
pastettes, and one plastic teaspoon. Turn on the UV light for
15 min and then turn it off before starting.
3. For each pup, fill a 100 × 15-mm sterile Petri dish with 5 mL of
ice-cold Dissecting Medium and place a polyethylene disc on
top of the cutting table of the chopper (see Note 9). Insert the
Whatman filter paper disc inside the inverted lid of the Petri
dish and place the two scissors, the curved tweezers, and the
teaspoon into a 100-mL beaker (filled to one-third with 70%
ethanol). Take the beaker and the Petri dish lid with the filter
paper to the animal house.
348 E. Gerace et al.
Fig. 1. How to remove the brain from a rat pup. After removing the skin (a), cut the skull bone with small scissors along the
sagittal middle line from back to front (b), ending with two diagonal cuts toward each eye (c). Remove the skull bone with
curved tweezers and, using a teaspoon, transfer the whole pup brain (d) to a filter paper placed on an inverted Petri dish lid.
4. Cut the head of a rat pup (see Note 10) with the large pair of
scissors. Use the small scissors to delicately remove the skin,
cut the skull bone along the sagittal middle line from back to
front, ending with two diagonal cuts toward each eye (see
Fig. 1). Remove the skull bone with the curved tweezers, and
using the teaspoon, transfer the whole pup brain to the filter
paper on the Petri dish lid.
5. Under the sterile hood, place the Petri dish lid with the brain
onto a piece of black cardboard and illuminate with the Schott
cold light source. Divide the two hemispheres with the scalpel
and, with the use of the paddle pastette, remove the cerebel-
lum, brainstem, and midbrain. Cut out the portion of the cor-
tex which is rostral to the hippocampus with the scalpel. Insert
the paddle pastette between the remaining cortex and the hip-
pocampus, raise and remove the hippocampus, and place it on
the polyethylene disc on top of the cutting table of the chop-
per (see Note 11).
6. Arrange the two hippocampi on the polyethylene disc, side by
side. Start the chopper (set at 420 μm) and cut the whole
length of both hippocampi. Remove the polyethylene disc with
the straight tweezers, turn it over, and dip it gently into the
Petri dish containing the 5 mL of ice-cold Dissecting Medium,
thus releasing the slices into the medium.
7. Allow gentle separation of the slices using a paddle pastette.
Take out the 6-well plates with the Millicell-®CM insert from
the incubator. Select the best slices (see Note 12) using the
stereomicroscope and place four of them very gently onto each
humidified insert using a paddle pastette. Place the plates in an
incubator at 37°C and 100% humidity in 95% air/5% CO2.
29 Rat Hippocampal Slice Culture Models… 349
3.2. Excitotoxicity Hippocampal slices cultured for 14 DIV retain an organotypic orga-
in Organotypic nization in which the pyramidal and granule cell layers can be clearly
Hippocampal Damage defined when observed under phase-contrast microscopy or follow-
ing toluidine blue staining (4, 7). Control slices incubated with PI
display very low fluorescence levels, but when cultures are exposed
to glutamate receptor agonists, PI staining increases dramatically.
Maximal damage is achieved in this system by exposing the slices to
10 mM glutamate for 24 h: Virtually all neuronal populations are
destroyed, and PI fluorescence in CA1 and CA3 increases several-
fold above background levels (see Fig. 2). In order to obtain the
layer-selective injury shown in Fig. 2, we use relatively low concen-
trations of NMDA (10 μM), AMPA (10 μM), or kainate (5 μM) for
24 h. It is also possible to expose the slices to higher concentrations
of NMDA (300 μM), AMPA (100 μM), or kainate (100 μM) for a
brief period of time (60 min), but in this case, the damage will be
more diffuse and less selective (see examples in 8). All slices used in
one experimental session are prepared from the same litter, i.e.,
from neonatal rat pups born on the same day:
1. Before the experiment, perform a “PI viability test” (see Note
13) by adding 6 μL of the 1-mg/mL stock solution of PI into
the wells containing the cultures slices (final concentration:
5 μg/mL). Rinse gently and observe under the fluorescence
microscope 30 min later. Cultures displaying distinct PI fluo-
rescence in the pyramidal cell layers are excluded. A mild PI
uptake is sometimes observed in the dorsal blade of the den-
tate gyrus, even in healthy slices.
2. Briefly rinse the slices and then incubate with 1.2-mL Serum-
Free Medium that has previously been prewarmed to 37°C.
3. Under the hood, add to each well the appropriate microliters
from the stock solutions (use intermediate dilutions if needed)
in order to reach the desired final concentration of glutamate
receptor agonists: 10 mM glutamate, 10 μM NMDA, 10 μM
AMPA, and 5 μM kainate. Treat control slices with Serum-
Free Medium.
4. Incubate for 24 h by placing the 6-well plates in the incubator
at 37°C and 100% humidity in 95% air/5% CO2 for recovery.
Neuronal injury is evaluated 24 h later.
3.3. OGD in To mimic the conditions that occur following cerebral ischemia
Organotypic in vivo, 14 DIV organotypic hippocampal slices are exposed to
Hippocampal Slices OGD in a hypoxia chamber for selected periods of time. Exposure
350 E. Gerace et al.
Fig. 2. Neuronal death induced by glutamate receptor agonists in rat organotypic hip-
pocampal slices. Cultured slices are exposed to drugs, incubated with PI (5 μg/mL), and
observed under fluorescence optics. Hippocampal slices under normoxic and drug-free
(control) conditions display background PI fluorescence. Hippocampal slices observed fol-
lowing 24 h exposure to 10 mM glutamate display a maximal degree of neuronal death in
all hippocampal layers. Hippocampal slices observed following 24 h exposure to 10 μM
NMDA display more intense fluorescence in the CA1 pyramidal cell layer, whereas follow-
ing exposure to 10 μM AMPA PI labeling is abundant in all layers. Following 24 h exposure
to 5 μM kainate fluorescence is more intense in the CA3 pyramidal cell layer.
29 Rat Hippocampal Slice Culture Models… 351
100
% of glutamate-induced PI fluorescence
**
80
**
60
20 min OGD
40
*
20
0 10 20 30
OGD duration (min)
30 min OGD
Fig. 3. Neuronal death induced by OGD in rat organotypic hippocampal slices. Cultured
slices are exposed to OGD for the indicated period and 24 h later incubated with PI for
fluorescence detection of its optical density in the CA1 region. OGD causes increasing
levels of CA1 injury when applied for 20–40 min. Data are expressed as percentage of
maximal damage produced by 24 h exposure to 10 mM glutamate. Hippocampal slices on
the left, photographed under fluorescence optics, display intense PI labeling in the CA1
subregion when exposed to increasing periods of OGD. Values represent the mean ± SEM
of at least five experiments performed in triplicate. *P < 0.05 and **P < 0.01 vs. basal PI
fluorescence (ANOVA + Tukey’s w test).
4. Notes
Acknowledgements
References
1. Gähwiler BH, Capogna M, Debanne D, (mGlu1) receptor antagonist 3-MATIDA
McKinney RA, Thompson SM (1997) reduces post-ischemic neuronal death.
Organotypic slice cultures: a technique has Neuropharmacology 42: 741–751
come of age. Trends Neurosci 20: 471–477 7. Pellegrini-Giampietro DE, Peruginelli F, Meli
2. Bahr BA (1995) Long-term hippocampal slices: E, Cozzi A, Albani-Torregrossa S, Pellicciari R,
a model system for investigating synaptic et al (1999) Protection with metabotropic glu-
mechanisms and pathologic processes. J tamate 1 receptor antagonists in models of
Neurosci Res 42: 294–305 ischemic neuronal death: time-course and
3. Moroni F, Meli E, Peruginelli F, Chiarugi A, mechanisms. Neuropharmacology 38:
Cozzi A, Picca R, et al (2001) Poly(ADP- 1607–1619
ribose) polymerase inhibitors attenuate necrotic 8. Werner CG, Scartabelli T, Pancani T, Landucci
but not apoptotic neuronal death in experi- E, Moroni F, Pellegrini-Giampietro DE (2007)
mental models of cerebral ischemia. Cell Death Differential role of mGlu1 and mGlu5 recep-
Differ 8: 921–932 tors in rat hippocampal slice models of ischemic
4. Pellegrini-Giampietro DE, Cozzi A, Peruginelli tolerance. Eur J Neurosci 25: 3597–3604
F, Leonardi P, Meli E, Pellicciari et al (1999) 9. Landucci E, Scartabelli T, Gerace E, Moroni F,
1-Aminoindan-1,5-dicarboxylic acid and Pellegrini-Giampietro DE (2011) CB1 recep-
(S)-(+)-2-(3 ¢ -carboxybicyclo(1.1.1)pentyl)- tors and post-ischemic brain damage: studies
glycine, two mGlu1 receptor-preferring antag- on the toxic and neuroprotective effects of can-
onists, reduce neuronal death in in vitro and nabinoids in rat organotypic hippocampal
in vivo models of cerebral ischemia. Eur J slices. Neuropharmacology 60: 674–682
Neurosci 11: 3637–3647 10. Pringle AK, Iannotti F, Wilde GJC, Chad JE,
5. Cozzi A, Meli E, Carlà V, Pellicciari R, Moroni Seeley PJ, Sundstrom LE (1997)
F, Pellegrini-Giampietro DE (2002) Neuroprotection by both NMDA and non-
Metabotropic glutamate 1 (mGlu1) receptor NMDA receptor antagonists in in vitro isch-
antagonists enhance GABAergic neurotrans- emia. Brain Res 755: 36–46
mission: a mechanism for the attenuation of 11. Abdel-Hamid KM and Tymianski M (1997)
post-ischemic injury and epileptiform activity? Mechanisms and effects of intracellular calcium
Neuropharmacology 43: 119–130 buffering on neuronal survival in organotypic
6. Moroni F, Attucci S, Cozzi A, Meli E, Picca R, hippocampal cultures exposed to anoxia/agly-
Scheideler MA, et al (2002) The novel and sys- cemia or to excitotoxins. J. Neurosci 17:
temically active metabotropic glutamate 1 3538–3553
Chapter 30
Abstract
Animal models of Parkinson’s disease are essential to explore pathophysiological hypotheses and to test
new treatment options, including neurotrophic factors. Catecholaminergic neurotoxins used to generate
such models are 6-hydroxydopamine and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. These neurotoxins
predominantly kill dopaminergic neurons through oxidative damage and mitochondrial failure, although
1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine fails to induce a significant dopaminergic neurodegeneration
in rats. The present chapter describes a protocol for the 6-hydroxydopamine rat model based on stereotaxic
injection performed only unilaterally, which mimics an early-to-mid stage of the disease.
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_30, © Springer Science+Business Media, LLC 2012
355
356 G. Mercanti et al.
2. Materials
2.1. Surgical 1. Kopf 900 stereotaxic instruments supplemented with ear bars
Apparatus and Tools (Kopf instruments).
2. 10-μL Hamilton syringe (Hamilton Bonaduz AG, Bonaduz,
GR, Switzerland).
3. Microinjection needles (27–30 gauge) connected to the
Hamilton syringe with a polyethylene tubing (PE 20, Becton
Dickinson) of about 25-cm length.
4. Surgical tools: scissors, sharp forceps, scalpels, sterile cotton
pads, syringes and needles, metal clips, suture clips and apply-
ing forceps, and ear punch (Fine Science Tools).
5. Electric shaver.
6. Dental cement.
7. Dremel 300 Series drill with engraving cutter 109 (Dremel
Italia).
8. Harvard compact infusion syringe pump (American Laboratory
Trading, Inc. East Lyme, CT, USA).
9. Heating pad for rodents and small animals (two Biological
Instruments, Besozzo, Italy).
3. Methods
3.1. Surgical All the animal procedures are carried out following guidelines
Procedures governing animal experimentation (European Union decree of
and Administration 24/11/1986 (86/609/IIC)) for the techniques described.
of 6-OHDA Before starting with the lesion, prepare all reagents and materials
that will be used. Once the animal is under anesthesia, everything
must be ready and near the stereotaxic instrument to avoid any
distraction from the surgical procedure.
1. Prepare a vial containing the solution of sterile 0.9% NaCl with
0.02% (w/v) ascorbic acid and put on ice.
2. Aliquot small amounts of approximately 1–2 mg of 6-OHDA-
HCl into Eppendorf® tubes and cover with aluminum foil to
avoid exposure to light. Store the aliquots at −20°C and avoid
repeated thawing/freezing. Be careful because 6-OHDA rap-
idly oxidizes when exposed to light and room temperature and
takes on a pinkish color.
3. Set up the surgical work station with all the surgical tools listed
above; disinfect with 70% ethanol.
4. Connect the needle with the PE tube to the Hamilton syringe.
Test by filling the syringe with the saline–ascorbic acid solution
and press it through the syringe and needle. This procedure
needs some practice and may seem time-consuming but is criti-
cal that there is no leakage from the PE tube or any occlusion
as this could prevent the toxin from reaching the target struc-
tures at the correct concentration (see Notes 1–3).
5. Place the needle on the stereotaxic frame and make sure that its
orientation is perfectly vertical and straight to avoid injection
on the wrong site.
6. Turn on the heating pad in such way that it is warm enough
when the animal will be placed in the stereotaxic frame.
7. Once everything is ready, weigh and anesthetize the animal.
Surgery can be performed using one of the different injectable
anesthesias proposed below:
● Fentanyl citrate salt 50 μg/mL + medetomidine HCl 50 μg/
mL, 20:1 mixture, 6.3 mL/kg intraperitoneal. To wake
30 A 6-Hydroxydopamine In Vivo Model of Parkinson’s Disease 359
Fig. 1. Stereotaxic surgery. Flat-skull positioning of the animal with ear bars (a) and tooth bar (b) on the stereotaxic frame
before starting surgery. (c) Shows the operatory field with skull bone exposure.
360 G. Mercanti et al.
Fig. 2. Stereotaxic coordinates. Dorsal and lateral views of rat skull. Antero/posterior (A/P) and lateral (L) coordinates are
referred to bregma point; dorsal/ventral (D/V) coordinates are calculated from the dural surface.
30 A 6-Hydroxydopamine In Vivo Model of Parkinson’s Disease 361
14. Retract the needle and drill a small hole in the skull bone above
the injection coordinate. Be careful not to rupture the
meninges when drilling (see Note 7).
15. Calculate the dorsal-ventral (DV) coordinate of the injection
having the dural surface as a reference plane, then make a small
hole in the meninges with either sharp forceps or an injection
needle. This will facilitate the injection and avoid damage to
the needle tip.
16. Dissolve 4 mg of 6-OHDA-HCl in 1 mL of sterile 0.9% NaCl
with 0.02% ascorbic acid. The concentration of 6-OHDA-HCl
in this solution should be 3.6 mg/mL (corresponding to
3.0 mg/mL free base 6-OHDA). Always keep the aliquots in
the freezer until use and check the color of the solution. Once
dissolved, keep on ice and in the dark because 6-OHDA is
light and temperature sensitive and rapidly oxidizes. Prepare a
fresh solution every 90 min, approximately.
17. Rinse the syringe with saline containing 0.02% ascorbic acid.
18. Fill the syringe with the 6-OHDA solution.
19. Place the Hamilton syringe on the pump, connect the needle
with the PE tube to the Hamilton syringe, fill the circuit with
the toxin solution, and set the infusion flow rate to 0.5 μL/min.
20. Slowly lower the injection needle down to the desired depth.
21. Start the infusion pump and pressure-eject the desired volume
of 6-OHDA solution at a maximum rate of 0.5 μL/min.
22. Leave the needle in place for 5 min before slowly retracting it
(see Fig. 3).
23. Empty the syringe and clean with two washes of ethanol and
one of saline containing 0.02% ascorbic acid.
24. Rehydrate the wound area with sterile water and close by
suturing.
25. Apply analgesic postoperatively and mark the tail or the ear
with a progressive identification number.
26. Inject 2 mL of saline subcutaneously to prevent dehydration
during recovery from anesthesia. For ethical reasons, analgesic
treatment should be administered to the animals before they
regain consciousness following anesthesia. The postoperative
analgesia consists of:
● Glucose solution (glucose 5%): 3 mL subcutaneously
● Steroidal anti-inflammatory (i.e., betamethasone disodium
phosphate 1.5 mg/mL, Bentelan®, Glaxo Wellcome):
100 μL intramuscular
● Nonsteroidal anti-inflammatory (i.e., indomethacin,
Liometacen®, Chiesi): 100 μL intramuscular
● Antibiotic (i.e., ceftriaxone, Rocefin® 1, Roche): 100 μL
subcutaneously
362 G. Mercanti et al.
Fig. 3. Schematic representation of lesion procedure. The toxin 6-OHDA-HCl is injected into the selected area by means of
a 27/30-G needle connected to a polyethylene tube and a Hamilton syringe. A precision infusion pump has a constant rate
of 0.5 μL/min. At the end of toxin infusion, the needle is left in place for additional 5 min, retracting it slowly.
3.3. Comments: Choice 6-OHDA does not cross the blood–brain barrier and therefore is
of Lesion Site classically injected into the SNpc and/or the MFB and produces a
massive, virtually complete lesion of nigral dopaminergic cell
bodies; SNpc neurons begin to die within the first 12 h of injection,
30 A 6-Hydroxydopamine In Vivo Model of Parkinson’s Disease 363
4. Notes
References
1. Dauer W, and Przedborski S (2003) Parkinson’s 10. Przedborski S, Jackson-Lewis V, Naini A,
disease: mechanisms and models. Neuron Jakowec M, Petzinger G, Miller R, Akram M
39:889–909 (2001) The parkinsonian toxin 1-methyl-4-
2. Recchia A, Rota D, Debetto P, Peroni D, phenyl-1,2,3,6-tetrahydropyridine (MPTP): a
Guidolin D, Negro A, Skaper SD and Giusti P technical review of its utility and safety. J Neuro-
(2008) Generation of alpha-synuclein-based chem 76:1265–1274
rat model of Parkinson’s disease. Neurobiol Dis 11. Olanow CW (1993) A radical hypothesis
30:8–18 for neurodegeneration. Trends Neurosci
3. Kordower JH, Emborg ME, Bloch J, Ma SY, 16:439–444
Chu Y, Leventhal L et al (2000) Neurode- 12. Jenner P (2008) Functional models of Parkinson’s
generation prevented by lentiviral vector delivery disease: a valuable tool in the development
of GDNF in primate models of Parkinson’s of novel therapies. Ann Neurol 64(Suppl
disease. Science 290:767–73 2):S16–S29
4. Gash DM, Zhang Z, Ovadia A, Cass WA, Yi A, 13. Ungerstedt U (1971) Adipsia and aphagia after
Simmerman L et al (1996) Functional recovery 6-hydroxydopamine induced degeneration of
in parkinsonian monkeys treated with GDNF. the nigro-striatal dopaminergic system. Acta
Nature 380:252–255 Physiol Scand Suppl 367:95–122
5. Biju K, Zhou Q, Li G, Imam SZ, Roberts JL, 14. Ungerstedt U (1968) 6-Hydroxy-dopamine
Morgan WW et al (2010) Macrophage- induced degeneration of central monoamine
mediated GDNF delivery protects against dop- neurons. Eur J Pharmacol 5:107–110
aminergic neurodegeneration: a therapeutic 15. Paxinos G, Watson C (1986) Rat Brain in
strategy for Parkinson’s disease. Mol Ther; Stereotaxic Coordinates San Diego, Academic
doi:10.1038/mt.2010.107 Press
6. Yasuda T and Mochizuki H (2010) Use of 16. Deumens R, Björklund A and Prickaerts J (2002)
growth factors for the treatment of Parkinson’s Modeling Parkinson’s disease in rats: an evalua-
disease. Expert Rev Neurother 10:915–924 tion of 6-OHDA lesions of the nigrostriatal
7. Laganiere J, Kells AP, Lai JT, Guschin D, pathway. Exp Neurol 175:303–317
Paschon DE, Meng X et al (2010) An engi- 17. Lee CS, Sauer H and Bjorklund A (1996)
neered zinc finger protein activator of the Dopaminergic neuronal degeneration and motor
endogenous glial cell line-derived neurotrophic impairments following axon terminal lesion
factor gene provides functional neuroprotection by intrastriatal 6-hydroxydopamine in the rat,
in a rat model of Parkinson’s disease. J Neurosci Neuroscience 72:641–653
30:16469–16474 18. Sindhu KM, Banerjee R, Senthilkumar KS,
8. Lambert CE, and Bondy SC (1989) Effects of Saravanan KS, Raju BC, Rao JM et al (2006)
MPTP, MPP+ and paraquat on mitochondrial Rats with unilateral median forebrain bundle,
potential and oxidative stress. Life Sci but not striatal or nigral, lesions by the
44:1277–1284 neurotoxins MPP + or rotenone display
9. Glinka Y, Gassen M, and Youdim MB (1997) differential sensitivity to amphetamine and
Mechanism of 6-hydroxydopamine neurotox- apomorphine. Pharmacol Biochem Behav
icity. J Neural Transm Suppl 50:55–66 84:321–329
Chapter 31
Abstract
Brain microdialysis is an analytical technique used for the dynamic monitoring of brain neurochemistry in
awake, freely moving animals. This technique requires the insertion of a small dialysis catheter, called a microdi-
alysis probe, into a specific brain region, and its perfusion with an artificial extracellular fluid. The
microdialysate samples, obtained from the probe outlet, can be analysed using high-performance liquid
chromatography with electrochemical detection for the quantification of oxidizable molecules recovered
from the extracellular space. In this chapter, we describe a protocol for performing a microdialysis setup and
experiment in freely moving rats and mice. Furthermore, the high-performance liquid chromatographic
determination of ascorbic acid, uric acid, catecholamines, indolamines and derivatives is described in detail.
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_31, © Springer Science+Business Media, LLC 2012
365
366 G. Bazzu et al.
2. Materials
2.2. Animals and 1. Animals: male Wistar rats weighing 300 g or 10-week-old
Stereotaxic Surgery C57BL/6 mice (weighing 30 g) from Charles River (Ballina,
Italy) can be used for in vivo microdialysis experiments. The ani-
mals must be maintained under standard animal care conditions
(12:12-h light/dark cycle, lights coming on at 7 a.m., room
temperature 21°C), with food and water ad libitum. Prior to the
start of any experiment, the health of the rodents has to be
assessed according to published guidelines (11). All procedures
need to be specifically licenced under the European Community
directive 86/609 included in national legislatures (i.e. Decreto
No. 116/1992 of the Ministry of Public Health in Italy).
2. Brain atlas for stereotaxic coordinates: the George Paxinos’ vol-
umes entitled “The Rat Brain in Stereotaxic Coordinates” (12)
and “The Mouse Brain in Stereotaxic Coordinates” (13) are
the reference books for setting up a microdialysis experiment
in rats and mice, respectively.
3. Stereotaxic apparatus: stereotaxic frame for small animals
(Model 900, David Kopf Instruments, UK) for rats equipped
with a mouse adaptor for mice (Kopf Mouse Adaptor from
2Biol, Varese, Italy). We recommend placing the stereotaxic
frame on a vibration-free marble table to increase precision
during probe implantation and to reduce brain damage.
4. Neurosurgery materials: alcohol-based permanent marker
(OHPen 96 fine, Stabilo, Germany), Betadine disinfectant
(Meda Pharma, Italy), sterile cotton wool, anatomical and sur-
gical tweezers, scalpel and scalpel blades, 27-G needles, 1- and
2.5-mL disposable syringes (for intraperitoneal (i.p.) injection
370 G. Bazzu et al.
2.4. HPLC 1. Alltech 426 HPLC pump (Alltech Srl, Milan, Italy)
2. Rheodyne injection valves (Alltech Srl) equipped with a 25-μL
loop
3. 50-μL Hamilton syringes (Alltech Srl)
4. Alltech Adsorbosphere™ C18 Column (100 × 4.6 mm;
Alltech Srl)
5. Antec EC controlled equipped with VT03 cell having a glassy
carbon working electrode, a Ag/AgCl reference electrode and
a stainless steel counter electrode (Alfatech Srl, Milan, Italy)
6. HPLC polyetheretherketone (PEEK) tubing and accessories
for 25-μL loop and connections (Alfatech Srl)
7. Varian data acquisition software (Varian Star v. 5, Varian Inc.,
USA) and hardware (Varian 800 MIB, Varian)
8. Personal computer (1 GB RAM and 250 GB hard disk)
equipped with an Ethernet board (operating system: Windows
XP/Vista/Windows 7)
31 Brain Microdialysis in Freely Moving Animals 371
2.5. Statistical Software for data storage, scientific graphing and statistical analysis:
Analysis Microsoft Excel 2007 (Microsoft Inc., USA), GraphPad Prism 5.0
(GraphPad Software, La Jolla, CA, USA), KaleidaGraph 4.0 (Synergy
software, Reading, PA, USA)
3. Methods
Fig. 1. Microdialysis probe construction procedure. See text for a detailed explanation of
the steps necessary for obtaining a working probe implantable in rat or mice striatum.
372 G. Bazzu et al.
tubing (see Fig. 1l). Gently remove the excess epoxy using a
clean 27-G needle. During this step, the membrane will be
shielded which allows the dialysis process only in the probe’s
active area (around 4 mm for rats and 2 mm for mice). Leave
the microdialysis probe to dry for at least 30 min at room
temperature.
15. Store the microdialysis probe in a fresh and dark environment,
far from dust and moisture, and keep it in place for at least
24 h before use to allow complete polymerization of the epoxy
resin.
3.2. Microdialysis (a) Cut one piece (200 mm in length) of PE50 tubing using a
Probe In Vitro razor blade.
Calibration (b) Cut a second piece of PE50 tubing (50 mm in length).
3.2.1. Circuit for In Vitro (c) Cut a 23-G needle (10 mm in length) using the drill mounting
Probe Calibration (see a diamond wheel.
Fig. 2) (d) Insert the 23-G needle (for 4 mm) inside the probe outlet.
(e) Fill the Hamilton syringe (1 mL) with the aECF (see Note 3).
(f) Connect one end of the 200 mm PE50 tubing to the Hamilton
syringe needle and the other end to the probe inlet.
(g) Connect one end of the 50 mm PE50 tubing to the 23-G
needle previously inserted in the probe outlet.
(h) Place the Hamilton syringe on the infusion pump and fill the
circuit using the maximum flow rate (see Note 4).
3.2.2. In Vitro Probe (a) Place the microdialysis probe on the stereotaxic frame holder
Calibration (see Fig. 2) and set the infusion pump flow rate to 1.5 μL/min.
(b) Unfreeze one Eppendorf vial, containing the calibration solu-
tion, to room temperature.
(c) Insert the probe membrane (hollow fibre) in the calibration
solution and wait 10 min to allow for stabilisation of the
microdialysis gradient.
(d) Insert the free end of the 50 mm PE50 tubing in a 250-μL
Eppendorf vial and wait 20 min.
(e) Remove the Eppendorf vial (containing 30 μL of dialysate)
and store at −80°C until HPLC analysis (see Subheading 3.5).
At the same time, take a 30 μL sample of the calibration solu-
tion (in which the probe is immersed) and store at −80°C.
Repeat this step two more times.
(f) After HPLC analysis, calculate the percent recovery of the
probe for every neurochemical component in the calibration
solution, using the following formula:
Recovery (%) = [microdialysate] / [calibration solution] × 100 (1)
Fig. 2. Circuit for the in vitro calibration of the microdialysis probe before in vivo use. The ratio between the concentrations
of neurochemicals, determined in the microdialysate and in the calibration solution, represents the probe recovery.
Fig. 3. Neurosurgery procedure used for implantation of the microdialysis probe in the striatum of anaesthetized rats or
mice. A stereotaxic frame for small animals is used for rats while it needs to be equipped with a mouse adaptor for mice.
See text for a detailed explanation of the steps illustrated in the figure.
376 G. Bazzu et al.
Fig. 4. Circuit for in vivo microdialysis in freely moving animals. The introduction of the liquid swivel prevents the formation
of sharp bends and folding of the polythene tubing during the animal’s rotations.
3.4. In Vivo (a) Cut two pieces (300 mm in length each) of PE50 tubing using
Microdialysis a razor blade.
3.4.1. Circuit for In Vivo (b) Cut a third piece of PE50 tubing (150 mm in length) and roll
Microdialysis Experiment up it to obtain three or four coils. Fix the coiled tubing with a
(see Fig. 4) small piece of adhesive tape as illustrated in Fig. 4.
(c) Cut a 23-G needle (10 mm in length) using the drill mounting
a diamond wheel.
(d) Insert the 23-G needle (for 4 mm) inside the probe outlet.
(e) Fill the Hamilton syringe (1 mL) with the aECF (see Note 3).
(f) Connect one end of the first 300 mm PE50 tubing to the
Hamilton syringe needle and the other end to the liquid swivel
inlet.
(g) Connect one end of the second 300 mm PE50 tubing to the
liquid swivel outlet and the other end to the probe inlet (the
probe is inserted in the brain).
(h) Place the Hamilton syringe on the infusion pump and fill the
circuit using the maximum flow rate (see Note 4).
3.4.2. In Vivo Experiment (a) Set the infusion pump flow rate to 1.5 μL/min and wait 60 min
with Freely Moving Animals to allow for stabilisation of the microdialysis gradient.
(see Fig. 4) (b) Connect one end of the 150 mm PE50 tubing to the 23-G
needle previously inserted in the probe outlet and wait 20 min.
31 Brain Microdialysis in Freely Moving Animals 377
(c) Remove the 150 mm PE50 tubing from the animal head dis-
connecting it from the 23-G needle attached to the probe
outlet.
(d) Aspirate the microdialysate present inside the 150 mm PE50
tubing using a 50-μL Hamilton syringe, transfer it (around
30 μL) to a 250-μL Eppendorf vial and store at −80°C (see
Note 9) until HPLC analysis (Subheading 3.5). Repeat steps
b–d for every microdialysis sample recovered during the in vivo
experiment.
(e) Usually three consecutive microdialysis samples are sufficient
for defining the baseline of the examined neurochemicals (see
Note 10).
(f) After baseline sample recovery and if desired, a pharmacologi-
cal treatment can be performed via systemic or intracerebral
drug administration. D-Amphetamine prepared as previously
described (Subheading 2.1, step 4) can be injected subcutane-
ously (2 mg/kg corresponding to 1.0 mL in rats and 100 μL
in mice) or by intracerebral retrodialysis. In the second case,
the aECF containing 2 mM D-amphetamine needs to replace
the perfusion fluid present in the microdialysis circuit (see Note 11)
during the entire period of treatment (usually comprising
between 20 and 60 min, corresponding to 1–3 microdialysis
samples). Immediately after the systemic drug administration
or during its intracerebral retrodialysis, the microdialysis sam-
ples are recovered as illustrated previously (steps b–d).
(g) After the end of the intracerebral drug administration, the solu-
tion containing only aECF has to be restored (see Note 11).
(h) After the pharmacological treatments, microdialysates can be
recovered for a period varying from 120 to 240 min (corre-
sponding to 3–6 samples).
(i) At the end of the experiment, disconnect the animal from the
microdialysis circuit and sacrifice it using the euthanasia solu-
tion (described in Subheading 2.1.3) injected intraperitoneum
(2.0 mL in rats or 200 μL in mice). At this point, you can
verify the correct positioning of the microdialysis probe as
described elsewhere (10).
3.5. HPLC Setup 1. As illustrated in Fig. 5, the mobile phase is moved by an iso-
and Sample Analysis cratic pump, through PE tubes, within the Rheodyne injector
equipped with a 25-μL loop. Through the injector, 30 μL
sample is loaded and channelled into the C18 column where
the substances are separated, revealed by electrochemical cell
and quantified by the proper software (14, 15).
2. One hour prior to starting the experiment, check the electro-
chemical cell for applied potential (+780 mV vs. Ag/AgCl ref-
erence electrode), then increase gradually the mobile phase
378 G. Bazzu et al.
Fig. 5. HPLC setup for ascorbic acid, uric acid, catecholamine and indolamine quantification in microdialysate samples
from the striatum of freely moving animals. The electrochemical detection (EC) allows the amperometric oxidation of the
abovementioned neurochemicals using a +780 mV potential vs. Ag/AgCl reference electrode. The circuit is completely
metal-free for preventing sample degradation during the analysis. WE glassy carbon working electrode; RE Ag/AgCl refer-
ence electrode; CE stainless steel counter electrode.
3.6. Microdialysis Data The concentrations in the dialysate will be expressed in nM (DA,
Analysis L-DOPA, NE, 3HT and 3MT) or μM (AA, UA, DOPAC, HVA
and 5-HIAA) and given as mean ± SEM. D-Amphetamine effects
on neurochemicals can be evaluated as absolute or percent changes
from the preceding baseline (for this purpose data need to be
expressed as percentage of baseline). Statistical significance is
assessed using analysis of variance (ANOVA) for differences over
time. Differences within groups are determined by paired t tests,
with the Newman-Keuls multiple comparison adjustment. Pear-
son’s correlation coefficient between neurochemicals (i.e. DA vs.
3-MT, AA vs. DA or DA vs. DOPAC + HVA) could be calculated
in each animal after drug administration.
4. Notes
Acknowledgments
References
1. Chaurasia CS, Müller M, Bashaw ED, Benfeldt of intracellular water space in humans. Am
E, Bolinder J, Bullock R, et al (2007) AAPS- J Physiol 253, 228–231
FDA Workshop White Paper: Microdialysis 6. Olson RJ, and Justice JB, Jr (1993) Quantitative
Principles, Application and Regulatory microdialysis under transient conditions. Anal
Perspectives Pharm Res 24, 1014–1025 Chem 65, 1017–1022
2. Delgado JMR, DeFeudis FV, Roth RH, Ryugo 7. Wang P, Wong S, and Sawchuk RJ (1993)
DK, and Mitruka BM (1972) Dialytrode for Microdialysis calibration using retrodialysis and
long-term intracerebral perfusion in awake mon- zero-net flux: application to a study of the dis-
keys. Arch Int Pharmacodyn Ther 198, 7–21 tribution of zidovudine to rabbit cerebrospinal
3. Ungerstedt U, and Pycock C (1974) Functional fluid and thalamus. Pharm Res 10, 1411–1419
correlates of dopamine neurotransmission. Bull 8. Charalambides C, Sgouros S, and Sakas D
Schweiz Akad Med Wiss 30, 44–55 (2010) Intracerebral microdialysis in children.
4. O’Neill RD, Rocchitta G, McMahon CP, Serra Childs Nerv Syst 26, 215–220
PA, and Lowry JP (2008) Designing sensitive 9. Nordström CH (2010) Cerebral energy metab-
and selective polymer/enzyme composite bio- olism and microdialysis in neurocritical care
sensors for brain monitoring in vivo. Trends Childs Nerv Syst 26, 465–472
Anal Chem 27, 78–88 10. Miele M, Mura MA, Enrico P, Esposito G,
5. Lönnroth P, Jansson PA, and Smith U (1987) A Serra PA, Migheli R, et al (2000) On the
microdialysis method allowing characterization mechanism of d-amphetamine-induced changes
31 Brain Microdialysis in Freely Moving Animals 381
in glutamate, ascorbic acid and uric acid release 14. Rocchitta G, Migheli R, Esposito G, Marchetti
in the striatum of freely moving rats. Br J B, Desole MS, Miele E, et al (2006) Endogenous
Pharmacol 129, 582–588 melatonin protects L-DOPA from autoxida-
11. Morton DB, and Griffiths PHM (1985) tion in the striatal extracellular compartment of
Guidelines on the recognition of pain, distress the freely moving rat: potential implication for
and discomfort in experimental animals and a long-term L-DOPA therapy in Parkinson’s dis-
hypothesis for assessment. Vet Rec 116, 431–436 ease. J Pineal Res 40, 204–213
12. Paxinos G, and Watson C (1986) The Rat 15. Rocchitta G, Migheli R, Mura MP, Grella G,
Brain in Stereotaxic Coordinates. Academic Esposito G, Marchetti B, et al (2005) Signaling
Press, San Diego pathways in the nitric oxide and iron-induced
13. Paxinos G, and Franklin KBJ (2001) The dopamine release in the striatum of freely mov-
Mouse Brain in Stereotaxic Coordinates Second ing rats: role of extracellular Ca2+ and L-type
Edition. Academic Press, San Diego Ca2+ channels. Brain Res 1047, 18–29
Chapter 32
Abstract
Neonatal sciatic nerve lesion is a useful experimental model for the study of neuronal cell death. Sciatic
nerve transection or crush is the most frequently used approach to evaluate motoneuron loss in the lumbar
enlargement of the spinal cord. Here we describe and illustrate the surgical procedures performed in our
laboratory to assess motoneuron cell death and the related cellular mechanisms.
Key words: Neonatal rat, Sciatic nerve, Transection, Crush, Motoneuron, Neuronal cell death
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_32, © Springer Science+Business Media, LLC 2012
383
384 A.S. Vieira et al.
proximal and distal, which are kept distant one from the other.
Therefore, since physical contact between the stumps is not allowed,
axonal regeneration is impaired (11). When performed at the first
postnatal day (P0), such type of injury determines the loss of approx-
imately 100% of the lesioned motoneurons in the lumbar enlarge-
ment of the spinal cord. Such extensive neuronal cell loss is not
achieved when the lesion is performed in older animals, particularly
adults. Besides disruption of trophic support from the target, it is
hypothesized that immature nonneuronal cells present in the periph-
eral nerves, especially Schwann cells, would not be able to provide
the lesioned motoneurons with trophic factors. The opposite would
be true for the sciatic nerve of adult animals (5, 12–14).
Another experimental model used to evaluate motoneuron
damage is sciatic nerve crush. Conversely to the nerve fiber transection
model, the axons are not totally sectioned. In fact, as neural fibers
are disrupted by compression, the nerve stumps are thought to
remain connected, which would allow for neuronal regeneration.
Specifically, cellular recovery would be favored by a microenviron-
ment established at the injury site, in which growth factors and
chemokines released by cells and molecules of the extracellular
matrix would allow remodeling of the nerve stumps, axonal recovery,
and neuronal regeneration, at least in part, after the lesion. However,
it is important to emphasize that Schwann cells of neonatal rats
have reduced competence to support regenerating motoneurons
when compared to adult Schwann cells. One hypothesis that may
be put forward is that immature Schwann cells have reduced
production of trophic factors (1, 5, 15, 16).
Motoneuron lesion induced by sciatic nerve transection or
crush in neonatal rats is an experimental model that can be used in
many research fields of neuroscience, such as neuronal cell death
pathways, peripheral nerve regeneration, and functional recovery
and evaluation of trophic factors and neuroprotective agents. In
our laboratory, we have focused on the use of exogenous growth
factors and neuroprotective substances on motoneuron survival
after sciatic transection in neonatal rats. In particular, we have been
investigating neuronal cell survival and the possible related cellular
pathways through motoneuron counting and evaluating protein
and gene expression (9, 10, 17, 18). Here we describe the surgical
procedures currently performed in our laboratory to induce
motoneuron injury in the lumbar spinal cord of neonatal rats by
either transectioning or crushing the sciatic nerve.
2. Materials
4. Microneedle holder
5. Scalpel #11
6. 8-0 suture silk
7. Glass Petri dish
8. Cotton swabs
9. 100-W incandescent lamp
10. Surgical microscope
3. Methods
3.1. Surgical 1. Separate rat pups from their mother in a small plastic box and
Procedures maintain them under a 100-W incandescent lamp until the
experiment is complete (see Note 1).
2. Immerse one rat in crushed ice for 4 min to induce anesthesia
by hypothermia (see Note 2).
386 A.S. Vieira et al.
Fig. 1. Anatomical location of the neonatal rat sciatic nerve as seen under the surgical microscope. (a) The approximate
location of the sciatic nerve (SN) is shown as a dotted line on the skin. The inset shows the region magnified in (a). (b, c)
The SN runs under the vastus lateralis (VL) and the adductor magnus (AM) muscles. (d, e) The VL and AM muscles were
removed to expose the SN. (f) Higher magnification of the SN. The arrow indicates the proximal region of the nerve. (c, d)
The nerve and the muscles are indicated by dotted lines.
Fig. 2. Anatomical location of the neonatal rat sciatic nerve as seen under the surgical microscope. (a) Incision on the
midthigh skin. (b) Exposed sciatic nerve after separation of the vastus lateralis and the adductor magnus muscles. (c) Higher
magnification of the region shown in (b). (d) The segment of nerve removed after transection is shown between the tips of
the microscissors. Bar: 3.0 mm (a, b, d), 1.2 mm (c).
10. Place the neonate rat under the 100-W incandescent lamp until
recovery is observed (see Note 5).
11. Return the pups to their mother (see Note 5).
3.2. Transcardiac 1. Separate rat pups from their mother in a small plastic box and
Perfusion maintain them under a 100-W incandescent lamp (see Note 1).
2. For anesthesia, administer the solution of ketamine (75 mg/kg)/
xylazine (15 mg/kg) intraperitoneally.
3. When the rat does not display any pedal reflex, fixate it on the
surgical table.
4. Make a skin incision through the midline, extending from the
sternal angle to the pubic region.
5. In the abdominal region, make a bilateral incision parallel to
the subcostal region and a midline linear incision. In this
procedure, the muscular and peritoneal membrane will be sec-
tioned, exposing the abdominal cavity.
388 A.S. Vieira et al.
3.3. Spinal Cord 1. After perfusion, fixate the rat in the prone position (decubitus
Dissection ventralis) on a surgical table.
2. Make an extensive dorsal skin incision through the midline to
expose the paravertebral musculature. From this moment on,
perform all procedures under the surgical microscope.
3. Gently remove the paravertebral musculature using a scalpel
and the vertebrae with the microforceps. Then, section the
meningeal covering to expose the spinal cord (see Note 8).
4. For motoneuron investigation, isolate the lumbar enlargement
from the remaining spinal cord using a scalpel. Remove the
lumbar enlargement from the spinal canal, sectioning the nerve
roots and meningeal membrane using microscissors.
5. The isolated lumbar enlargement may be processed using stan-
dard histological protocols for paraffin embedding or frozen
processing (see Note 9).
4. Notes
Fig. 3. Transverse sections of the lumbar enlargement (L4) of rats 5 days after sciatic transection performed at an age of 2
days postnatally. (a) Reduction of the number of motor neurons is observed in the right ventrolateral region (shown in
(c)) as compared to the contralateral unlesioned side (shown in (b)). Paraffin-embedded sections stained with cresyl violet.
Bar: 500 μm (a), 100 μm (b, c).
References
1. Kuno M (1990) Target dependence of the degeneration of motor neurons after
motoneural survival: the current status. J Neurosci axotomy. Nature 345, 440–441
Res 9, 155–172 5. Lowrie MB, and Vrbova G (1992) Dependence of
2. Oppenheim RW (1991) Cell death during postnatal motoneurones on their targets: review
development of the nervous system. Ann Rev and hypothesis. Trends Neurosci 15, 80–84
Neurosci 14, 453–501 6. Sendtner M, Holtmann B, Kolbeck R, Thoenen
3. Levi-Montalcini, R (1964) The nerve growth H, and Barde YA (1992) Brain-derived neu-
factor. Ann NY Acad Sci 118, 149–170 rotrophic factor prevents the death of motoneu-
4. Sendtner M, Kreutzberg GW, and Thoenen H rons in newborn rats after nerve section.
(1990) Ciliary neurotrophic factor prevents Nature 360, 757–759
32 Evaluating Motor Neuron Death in Neonatal Rats Subjected… 391
7. Greensmith L, and Vrbova G (1996) neurotrophic factor in the sciatic nerve of the
Motoneurone survival: a functional approach. adult rat after lesion and during regeneration.
Trends Neurosci 19, 450–455 J Cell Biol 118, 139–148
8. Oliveira AL, Risling M, Negro A, Langone F, 16. Magill CK, Moore AM, Yan Y, Tong AY,
and Cullheim S (2002) Apoptosis of spinal MacEwan MR, Yee A, et al. (2010) The dif-
interneurons induced by sciatic nerve axotomy ferential effects of pathway- versus target-
in the neonatal rat is counteracted by nerve derived glial cell line-derived neurotrophic
growth factor and ciliary neurotrophic factor. factor on peripheral ner ve regeneration.
J Comp Neurol 447, 381–393 J Neurosurg 113, 102–109
9. Rezende ACS, Vieira AS, Rogerio F, Rezende 17. Rogerio F, De Souza Queiroz L, Teixeira SA,
LF, Boschero AC, Negro A, et al. (2008) Effects Oliveira AL, De Nucci G, and Langone F
of systemic administration of ciliary neurotrophic (2002) Neuroprotective action of melatonin
factor on Bax and Bcl-2 proteins in lumbar spinal on neonatal rat motoneurons after sciatic nerve
cord of neonatal rats after sciatic nerve transec- transection. Brain Res 926, 33–41
tion. Braz J Med Biol Res 41, 1024–1028 18. Rogerio F, Teixeira SA, Rezende ACS, Cofino
10. Rezende ACS, Peroni D, Vieira AS, Rogerio F, R, Queiroz LS, De Nucci G, et al. (2005)
Talaisys RL, Costa FTM, et al. (2009) Ciliary Superoxide dismutase isoforms 1 and 2 in lum-
neurotrophic factor fused to a protein trans- bar spinal cord of neonatal rats after sciatic
duction domain retains full neuroprotective nerve transection and melatonin treatment.
activity in the absence of cytokine-like side Dev Brain Res 154, 217–225
effects. J Neurochem 109, 1680–1690 19. Phifer CB, and Terry LM (1986) Use of hypo-
11. Vejsada R, Sagot Y, and Kato AC (1995) thermia for general anesthesia in preweanling
Quantitative comparison of the transient res- rodents. Physiol Behav 38, 887–890
cue effects of neurotrophic factors on axoto- 20. Cammermeyer J (1961) The importance of
mized motoneurons in vivo. Eur J Neurosci 7, avoiding “dark” neurons in experimental neu-
108–115 ropathology. Acta Neuropathol 1, 245–270
12. Schmalbruch H (1984) Motoneuron death 21. West MJ, Slomianka L, and Gundersen
after sciatic nerve section in newborn rats. J Comp HJG (1991) Unbiased stereological esti-
Neurol 224, 252–258 mation of the total number of neurons in
13. Schmalbruch H (1987) Loss of sensory neu- the subdivisions of the rat hippocampus
rons after sciatic nerve section in the rat. Anat using the optical fractionator. Anat Rec
Rec 219, 323–329 231, 482–497
14. Lowrie MB, Lavalette D, and Davies CE (1994) 22. Gundersen HJ, Bagger P, Bendtsen TF, Evans
Time course of motoneurone death after neo- SM, Korbo L, Marcussen N, et al. (1988) The
natal sciatic nerve crush in the rat. Dev Neurosci new stereological tools: dissector, fractionator,
16, 279–284 nucleator and point sampled intercepts and
15. Sendtner M, Stöckli KA, and Thoenen H their use in pathological research and diagnosis.
(1992) Synthesis and localization of ciliary APMIS 96, 857–881
Chapter 33
Abstract
Spinal cord injury (SCI) is a serious clinical problem that causes lifetime disabilities to victims and inflicting
huge social burden on our society. One of the main lacunae in developing potential therapeutic measures
in SCI is a lack of suitable animal models that could be comparable to clinical situations. Thus, develop-
ment of new animal models of SCI is highly needed to expand our knowledge on cell injury and repair
process in order to reduce cord pathology, and in translating advanced therapies in patients of SCI to
improve therapeutic strategies. Keeping these views in mind, a suitable animal model is developed in our
laboratory that can be used to explore new therapeutic tools in SCI. The details of our methods used to
induce SCI in rodents and neuroprotection achieved by use of selected neurotrophic factors are described
in this chapter.
Key words: Spinal cord injury, New model, Neurotrophic factors, Neuroprotection, Cell injury,
Neurorepair
1. Introduction
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7_33, © Springer Science+Business Media, LLC 2012
393
394 H.S. Sharma and A. Sharma
and neuronal, glial, and myelin damage (1, 3–5). These changes
will continue further with advancement of time beyond the injury
point across the spinal cord in rostrocaudal directions leading to
permanent disability (1, 4, 5, 7). Thus, a detailed knowledge on
early pathological events is necessary in exploring suitable
therapeutic strategies to treat SCI victims.
Recent studies show that treatment schedule initiated after 3–6 h
of primary insult is largely ineffective on the spinal cord pathology
(1, 4, 5). Thus, studies using early therapeutic interventions to
reduce spinal cord cell and tissue damage are needed. Obviously,
when spinal cord neurons along with nonneuronal cells, e.g., glial
cells, endothelial cells, and myelin, are protected after injury, then
only neurorecovery and/or neuroprotection could be achieved
(1, 8–12). Restoration of spinal cord cell and tissue function after
trauma is crucial to reduce sensory motor disturbances (1, 2, 13–17).
Accordingly, neuroregeneration and neurorepair mechanisms appear
to be the most important factors for any future spinal cord thera-
peutic strategies after injury.
Our studies have shown that topical application of neurotro-
phins, including nerve growth factor (NGF), brain derived neu-
rotrophic factor (BDNF), or other growth factors, e.g., glial-derived
neurotrophic factor (GDNF) and ciliary neurotrophic factor (CTNF),
reduces spinal cord pathology and functional disabilities in a rat
model of SCI when administered repeatedly within 10–30 min
after trauma (5, 6, 9–12). This suggests that neurotrophins could
be the potential and ideal candidates for effective therapeutic strat-
egies in SCI. However, further studies are needed to understand
the potential beneficial effects of various neurotrophins or other
compounds in combination to effectively reduce the pathological
consequences of SCI. For this purpose, an effective animal model
of SCI is needed to explore neuroprotective efficacy of various
compounds in the vicinity of the lesion site as well in the ipsilateral
or contralateral side of the cord (13, 14, 16, 17). However, to date,
there are no models of SCI in which this ipsi- or contralateral
aspects of spinal cord pathology can be examined in detail. This is
largely due to the fact that most models include compression,
weight drop, or transaction, leaving no scope to study the effects
of lesion or trauma around the injury site (1, 3–5, 15). Thus, effects
of drugs in the ipsilateral or contralateral cord are still lacking in
other models of SCI.
In this chapter, we describe a new model of SCI in rats and
mice in which both ipsilateral and contralateral cell changes can
be examined within the lesion site as well as in the rostrocaudal
directions of the cord after trauma. These features make this
model unique in expanding our understanding of the pathophysi-
ological consequences of trauma within the spinal cord in a very
precise manner.
33 Rodent Spinal Cord Injury Model and Application of Neurotrophic… 395
2. Materials
2.3. Lab Apparatus 1. Liston bone cutter, straight: 14 cm/5.5 in. (Harvard Surgical
and Other Supplies Instruments)
2. Friedman Rongeur, curved: 14 cm/5.5 in. (Harvard Surgical
Instruments)
3. Dissecting stereomicroscope (Carl Zeiss, 20–30×)
4. Magnifying glass lamp (Magnifying Lamp 900-061).
5. Circular 22 W fluorescent light (Chicago electric)
6. Dual Lab standard stereotaxic frame for rats or mice, with 45°
ear bars (Harvard Apparatus)
7. EdgeAhead crescent knife, 1.75 mm edge length, 60°, bevel up
8. Gelco sponge (London)
9. PE 10 cannula
10. 21- and 36-gauge needles, butterfly needles
11. 50-mL-capacity Hamilton microliter syringes (HAMILTON
Bonaduz AG, Bonaduz, Switzerland)
12. Precision microinfusion pump (Razel Scientific Instrument
Inc, Stamford, CT, USA)
13. 3-in. Gamma counter (Packard)
14. Precision electronic balance (e.g., METTLER TOLEDO)
15. Oven capable of reaching 90°C (e.g., Class 100 Cleanroom
Ovens, Terra Universal)
16. Light microscope (Zeiss or similar type)
17. Ultramicrotome with diamond knife (LKB or comparable
supplier)
18. Phillips 400 transmission electron microscope (or comparable
model)
3. Methods
Table 1
Animal vs. human SCI factors, variability, and differences
Clinical Experimental
Multifactorial Simple
Flexion, extension, rotation, compression Weight drop, incision,a compression, transaction,
concussion, contusion, laceration hemisection, electrolytic lesion, contusion, clip
compression
Complex forces are active at the time of injury Only one injury factor is present
Closed vertebral system injury Open vertebral system injury
After laminectomy
Usually anterior compression Mainly posterior compression
Mainly occurs during state Produced under anesthesia conscious
Unlimited or uncontrolled time interval between Carefully controlled time schedule hospital
injury and first hand examination
Fig. 1. Diagrammatic representation of spinal cord injury (SCI) in the rat and sampling of spinal cord tissues T9 or T12 for
morphological or biochemical analyses (a). (b) Depth of lesion (L) in the right dorsal horn and selection of tissue areas from
the ipsi- or contralateral cord in dorsal horn (1) or ventral horn (2) is clearly identified. Bar = 5 mm. Data modified after
Sharma (4, 5, 9, 20).
33 Rodent Spinal Cord Injury Model and Application of Neurotrophic… 399
Table 2
Advantages and shortcomings of various experimental models of spinal cord injury
3.1. Animal Handling 1. Use rats or mice of specific age, sex, and strain according to
protocol. We used male Sprague–Dawley rats (180–250 g) or
C57 mice (30–35 g) from Alab, Stockholm, Sweden.
2. Keep the animals under controlled laboratory room temperature
(21 ± 1°C) with 12-h light and 12-h dark schedule, with food
pellets and tap water supplied ad libitum up to 6–7 days before
experiments.
400 H.S. Sharma and A. Sharma
3.3. Administration 1 Hold rats/mice firmly from the back skin using a towel, and
of Anesthesia take care that the animal is not able to move its head otherwise
you may get bitten. While holding the animal, firmly turn it
around to expose its belly toward you.
2 Use an already prepared anesthetic in a syringe (for dose and
choice of anesthesia, see below). Gently administer the anesthetic
into the peritoneal space that can be reached from the lower
groin with a needle angle of 30° and pushing about 0.5–1 cm
inside using a 36-G stainless needle. Just before injection, pull
the plunger toward you to see that the needle is inside the
peritoneal cavity. In such case, a vacuum is created (18, 19)
(see Note 2).
3 If the syringe appears tight and pulling the plunger results in a
vacuum, then push the correct dose of anesthesia slowly within
20–30 s.
4 After completion of the injection, gently return the rat to its
cage and watch for the effects of anesthetics. The rat will show
signs of anesthesia and will gradually fall down on the cage floor
and go to sleep. Use a mild tail pinch or ear pinch to check
the stage of anesthesia. If the animal does not respond to these
stimuli, it is in the surgical grade of anesthesia (Grade IV) (18).
3.4. Exposing 1. After surgical grade anesthesia is obtained, shave hairs on the
the Spinal Column back skin gently and expose the skin area covering the spinal
column over the thoracic region. Wash the skin surface with
70% alcohol to sterilize the skin surface.
402 H.S. Sharma and A. Sharma
3.5. Exposing 1. After exposing the T10–11 spines and articular processes,
the Spinal Cord insert one tip of a very-fine-pointed side-cutting bone cutter
(Laminectomy) carefully between T10 and T11 vertebral space.
2. Carefully remove the vertebral bone using space between the
two adjacent vertebrae. When you are able to perform on
the right side, use the same procedure to remove a part of the
vertebra on the left side as well. After that, use a precision
bone rongeur to widen the space by removing pieces of bones
slowly (see Note 6).
3. During the bone removal process, apply cold saline (4°C) over
the bone and spinal cord surface frequently to keep the tissue
wet all the time (16, 17).
4. Use a dissecting stereomicroscope to remove bone and to
avoid direct damage to the spinal cord, if necessary. However,
after some experience, a good magnifying glass lamp with a
circular 22 W fluorescent light will be enough to perform
laminectomy.
3.6. Making the Lesion 1. After laminectomy, stabilize the spinal column of the
rat or mice under a stereotaxic apparatus. Use constant
cold saline application over the exposed spinal cord.
2. Using one of the vertical axis manipulators of the stereotaxic
apparatus, fix the microknife with a straight bevel positioned
at 90° over one end of the exposed spinal cord (see Note 7).
3. Adjust the coordinates to fit the knifepoint over the right
dorsal horn (about 0.8 mm right to the dorsal spinal artery) of
the T10 segment. Carefully lower the tip of microknife to
33 Rodent Spinal Cord Injury Model and Application of Neurotrophic… 403
touch the dura over the right dorsal horn without piercing it.
Any accidental piercing will result in leakage of spinal fluid.
4. After positioning the knife over the right dorsal horn, insert
the tip of the knife within the spinal cord up to 1.5 mm deep
to reach around the Rexed laminae VIII to IX ((13–15, 17);
see Fig. 1).
5. After reaching deeper into the desired area, gradually move the
knife using stereotaxic apparatus longitudinally backward up to
5 mm (13–17). After the lesion, remove the knife from the
spinal cord immediately and cover the wound with cotton soaked
in saline to prevent drying of the cord tissue. If there is any
bleeding, gently soak it with Gelco sponge (see Fig. 1; Note 8).
3.6.1. Alternative Methods 1. An experienced worker could also make a lesion using a #11
to Induce Spinal Cord sterile carbon steel surgical blade by hand under a magnifying
Injury fluorescent lamp in rats or in mice (13).
2. For making lesion by hand, the knife blade may be sealed with
araldite leaving only 1.5 mm tip open to avoid accidental
injury due to spinal reflex while making incision. The knife
preparation can be done one night before the experiment (13, 16)
(see Note 9).
3.7. Postoperative 1. After the lesion, remove the animals from the stereotaxic plat-
Care form and place them in a secure plastic cage on good surgical
bedding. Do not put spinal cord injured animals in the cage
with wooden scrap as small pieces of wood may choke these
anesthetized rats/mice causing to death.
2. Place each rat/mice in individual cages to avoid any possible
attack by other animal or laceration of the wound in case ani-
mals need to be revived for behavioral investigations. However,
for the study of spinal cord pathophysiology, it is not necessary
to awaken them. In such cases, maintain the anesthetic dose
for the entire survival period as needed by the experimental
protocol (19).
3.8.1. Topical Application 1. Prepare the desired dilution of each neurotrophic factors in sep-
of Neurotrophins over arate 50-mL-capacity Hamilton microliter syringes. Connect the
the Spinal Cord syringe to a precision microinfusion pump for slow infusion.
Calibrate the speed of injection to deliver 10 mL/min to allow
0.25 mL over the spinal cord within 15 s (20, 21, 23, 24). Should
you choose for coapplication, replace with another neurotro-
phin-filled syringe after 15 s. Normally, a combination of 2–3
neurotrophic factors may be completed within less than a minute.
404 H.S. Sharma and A. Sharma
3.8.2. Selection of Dosage 1. In one group of experiments, apply BDNF or IGF-1 in separate
and Combination of group of rats (0.1 mg/10 mL in phosphate-buffered saline)
Neurotrophic Factors in SCI repeatedly, starting from 30 min before injury followed by 10,
30, and 60 min after injury. This application may continue
then every hour until 240 min after injury (27–30).
2. In other group of animals, apply various combinations of
BDNF with GDNF, NGF, NT-3 or IGF-1, or GDNF starting
from 30 to 90 min after SCI (or according to your plan) and
allow the animals to survive the desired time after injury. You
can control the total dose of the neurotrophins adjusted
(0.5 mg each) in 10–30 mL solution (0.5 mg of BDNF + 0.5 mg
of GDNF, or IGF-1, NGF or NT-3) and apply topically over
the injured spinal cord within 10 s (20, 21, 23, 24). Control
group may receive 10–30 m L of 0.9% saline instead of
neurotrophins (25–28).
3.9. Parameters Spinal cord injury induces rapid leakage of proteins across the
for Assessing BSCB and will cause edema and cell injury. These cell and tissue
Neuroprotection injuries progress over time across the longitudinal and transverse
axis of the spinal cord, leading to functional disability. It is likely
that neurotrophic factors if applied soon after SCI will reduce
BSCB disturbances, edema formation, and cell injury (20, 29, 30).
The following subsections describe parameters, which may be used
to assess the neuroprotective effects of neurotrophins on SCI.
3.9.1. BSCB Permeability To measure BSCB breakdown in spinal cord injury, we use Evans
blue and radioiodine ([131]-Iodine) as tracers. These tracers when
introduced into the circulation bind to serum proteins. Thus, their
leakage within the cord represents tracer–protein complex indicating
vasogenic edema formation (18, 19).
33 Rodent Spinal Cord Injury Model and Application of Neurotrophic… 405
3.9.2. Spinal Cord Edema Edema in the spinal cord could be determined by measuring water
Formation content of different spinal cord segments.
1. At the end of the experiment, remove the spinal cord and
divide it quickly into several segments such as T9, T10, T11,
and T12 and weigh them accurately using a precision
electronic balance (see Note 12).
2. Immediately after taking wet weight of the samples, place them
in an oven maintained at 90°C for 72 h. At this time, total
water may be evaporated and you could take the dry weight of
the sample (see Note 13).
3.9.3. Spinal Cord 1. Perfuse the rats or mice through the heart with either cold
Morphology 0.9% sterile saline (as mentioned above) or using cold phosphate-
buffered saline (0.1 M, pH 7.0) to washout blood inside the micro-
vessels (about 100–150 mL for rat and 60–80 mL for mice).
Table 3
406
Postinjury treatment with neurotrophins either alone or in combination on spinal cord injury (SCI) induced blood–spinal
cord barrier (BSCB) permeability, spinal cord edema formation, and cell injury in the T9 segment in rats. The SCI was
performed by making a longitudinal incision into the right dorsal horn of the T10–11 segments, and the animals were
allowed to survive 5 h after trauma [for details see text]
BDNF GDNF NT-3 BDNF + GDNF BDNF + GDNF BDNF + NT-3 BDNF + NT-3
Parameters
measured Control 5 h SCI +30 min +30 min +30 min +60 min +90 min 60 min 90 min
H.S. Sharma and A. Sharma
BSCB permeability
Evans blue 0.24 ± 0.04 1.65 ± 0.12** 0.87 ± 0.32b 0.79 ± 0.22c 1.67 ± 0.13ns. 0.72 ± 0.18c 0.89 ± 0.23b 1.68 ± 0.34** 1.73 ± 0.14**
(mg %)
[131]
Iodine (%) 0.35 ± 0.06 1.96 ± 0.14** 0.94 ± 0.12c 0.89 ± 0.12c 1.88 ± 0.26ns 0.81 ± 0.16c 1.07 ± 0.24b 1.98 ± 0.44** 2.08 ± 0.51**
Edema formation
Water content 66.12 ± 0.18 69.34 ± 0.23** 67.67 ± 0.21b 67.23 ± 0.18b 68.86 ± 0.34ns 67.16 ± 0.12c 68.21 ± 0.12b 69.84 ± 0.13** 69.98 ± 0.43**
(%)
Cell injury
Neuronal Nil 4 2 ± 1d 2 ± 1d 4 ± 1ns 2 ± 2d 2 ± 2d 4±1 4±1
damage
Endothelium Nil 4 2 ± 1d 2 ± 2d 4±1 2 ± 1d 3 ± 1d 3 ± 1d 4±1
damage
(La3+)
Data modified after Sharma (5–8) (see text)
Values are mean ± SD of 5–6 rats in each group
*P < 0.05; **P < 0.01 (compared from control)
a
BDNF, GDNF, or NT-3 (Total amount, 1 mg in 10 mL) was applied topically in separate group of animals after SCI. In combination, BDNF, GDNF, or
NT-3 (0.5 mg each, total dose 1 mg) was used in identical manner
b
P < 0.05
c
P < 0.01 (compared from 5 h SCI), ANOVA followed by Dunnett’s test from one control
d
P < 0.05, Chi-square test from 5 h SCI group, ns not significant (from 5 h SCI group)
33 Rodent Spinal Cord Injury Model and Application of Neurotrophic… 407
3.9.4. Ultrastructural 1. Perfuse with Somogyi fixative for better preservation of cell
Changes in the Spinal Cord membrane and nuclei (22).
2. To study BSCB breakdown at the ultrastructural level, add lan-
thanum chloride (2.5%, w/v) into the fixative before perfu-
sion. Lanthanum is an ion that can be seen at the ultrastructural
level across the microvessels without any special processing of
the tissue (25–28).
3. After perfusion, cut small tissue pieces from the desired spinal
cord segments and postfix them in osmium tetroxide and then
embed in araldite or electron microscope grade Epon 812.
4. Cut semithin sections (1 mm thick) from each block on a
microtome using a glass knife and stain them with toluidine
blue. Analyze these sections under a microscope and identify
special areas of the cord for ultrastructural observations (14,
15) (also see Fig. 2).
5. After identifying the desired areas of the spinal cord, trim the
block and cut ultrathin sections on an ultramicrotome using
diamond knife. Collect these ultrathin sections on a one whole
copper grid (or mesh grid) and counterstain them with uranyl
acetate and lead citrate before viewing them under a Phillips
400 transmission electron microscope.
6. Save the images using a digital camera using a basic magnification
of 4,000–6,000 of the grid for comparison (see ref. (4, 5)).
Lanthanum can be seen as crystallized dark black structures
around the microvessels and in the neuropil if BSCB break-
down occurred (4, 5) (see Fig. 3).
Fig. 3. Repeated application of BDNF or IGF-1 over spinal cord following injury induces neuroprotection. (Aa) BDNF or IGF-1
was applied (1 mg in 10 mL) over the spinal cord starting from 30 min before injury followed by 5, 10, 15, and 30 min after
trauma, and thereafter, 60, 120, 180, and 240 min after injury. The animals were allowed to survive 5 h (a). BDNF or IGF-1
reduced BSCB breakdown to radiotracer in different segments (b) that coincided well with reduction in edema formation
(c). Morphological investigations showed preservation of myelin as seen using myelin basic protein (MBP) immunoreactiv-
ity (Bb) as compared to untreated spinal cord MBP (Ba). Ultrastructural studies showed exudation of lanthanum into the
spinal cord neuropil and across the microvessels (arrows Bc) that was significantly reduced by BDNF treatment (Bd).
Bar: B:a,b = 50 mm, B:c.d: 1 mm. Data modified after Sharma (4, 5, 9).
410 H.S. Sharma and A. Sharma
4. Notes
Fig. 4. A suitable combination of neurotrophic factors is more effective than their treatment
alone. Thus, treatment with BDNF, IGF-1, and GDNF in combination reduces motoneuron
damage after trauma markedly (c) as compared to untreated injured group (d). Whereas,
a combination of BDNF, NGF, and NT-3 was not that effective in reducing cell damage after
SCI either given 60 or 90 min after primary insult (a, b). Bar = 40 mm. Data modified after
Sharma (4, 5, 9, 20).
6. Always use the bone rongeur moving outward to cut the bone
in order to avoid direct contact of metal parts with the spinal
cord. Try to make space over the dorsal spinal cord (about
5–6 mm), keeping the dorsal spinal artery in the middle. Avoid
any deep lateral opening over the spinal cord or spinal roots.
Also, make special care not to damage the dura matter while
removing the vertebral bone over the cord (6).
7. The Crescent microsurgery knife can easily penetrate tissue
without making injuries, and a precision lesion could be
produced without any laceration of the cord.
8. Take special care about the possible spinal reflex when any
metal touches the spinal cord. For this purpose, stabilization of
rat spinal cord is necessary by suitable clamps or using a firm
hold by hand over the back of the rat/mouse as and when
appropriate to avoid spinal reflex while making the injury (1, 4,
5). Normally, under surgical grade of anesthesia, spinal reflex
will be of minor degree during the lesion procedure. Any
unwanted movement of spinal cord due to spinal reflex will
damage spinal cord to different extents and result in variability
of data collection. If this happens, discard those animals from
data analyses.
9. Our laboratory compared results of spinal cord edema forma-
tion in rats and mice after microknife incision using stereotaxic
apparatus and free-hand lesion using scalpel blade (16). No
significant differences between handmade lesion and stereotaxic
apparatus-induced SCI could be noticed with regard to edema
formation or cell changes in the cord (16–19).
10. Repeated topical application of neurotrophins (0.1 mg–0.25 mg
/10 mL in phosphate-buffered saline) could gain rapid access
within the normal or injured cord tissue. Select the timing of
topical application starting from 10 to 30 min before injury
or to selected intervals after trauma ranging from 5 to 120 min
after the initial insult. An early manipulation of the spinal
cord after injury could result in thwarting spinal cord cell and
tissue injury and limit autodestructive changes in the cord.
11. Immediately before saline perfusion, take about 1 mL whole
blood sample after cardiac puncture and store it for analyses
of blood radioactivity or Evans blue concentration at the
time of killing.
12. The average wet weight of spinal cord segments could vary
from 60 to 90 mg in normal rats. Trauma to the cord may
increase the wet weight of the spinal cord samples from the
identical segments. Before weighing spinal cord injury samples,
remove large blood vessels or blood clots or hemorrhagic spots
over the injury site, as their presence will increase the wet
weight erroneously (19).
33 Rodent Spinal Cord Injury Model and Application of Neurotrophic… 413
13. If in doubt, take dry weight at least three times after an interval
of 4–6 h. When the dry weight in the last three measurements
became stable, use this weight to calculate the water based on
the differences between wet and dry weight of these samples
(for details see ref. (14–16)).
14. For good fixation of spinal cord tissues, wrap the perfused
animals separately in aluminum foil and keep it at 4°C for
overnight. On the next day, remove the spinal cord and place
the samples in the same fixative at 4°C for 3–4 days.
15. If the images require processing using Photoshop or any
other image analysis program, always use identical filter or
color processing on the computer for control and experi-
mental samples (31).
Acknowledgments
References
1. Schwab ME, Bartholdi D (1996) Degeneration 5. Sharma HS (2008) New perspectives for the
and regeneration of axons in the lesioned spinal treatment options in spinal cord injury. Expert
cord. Phys Rev 76, 319–370 Opin Pharmacother 16, 2773–2800
2. Stålberg E, Sharma HS, Olsson Y (1998) Spinal 6. Sharma HS (2004) Pathophysiology of the
Cord Monitoring. Basic Principles, blood-spinal cord barrier in traumatic injury.
Regeneration, Pathophysiology and Clinical In: HS Sharma and J Westman (Eds) The
Aspects, Springer, Wien, New York, pp 1–527 Blood-Spinal Cord and Brain Barriers in Health
3. Sharma HS, Westman J (2004) The Blood- and Disease, Elsevier Academic Press, San
Spinal Cord and Brain Barriers in Health and Diego, pp. 437–518
Disease, Academic Press, San Diego, pp. 1–617 7. Winkler T, Sharma HS, Stålberg E, Westman
(Release date: Nov. 9, 2003) (1998) Spinal cord bioelectrical activity, edema
4. Sharma HS (2011) Early microvascular reac- and cell injury following a focal trauma to the
tions and blood-spinal cord barrier disruption spinal cord. An experimental study using phar-
are instrumental in pathophysiology of spinal macological and morphological approaches.
cord injury and repair: novel therapeutic strate- In: Spinal Cord Monitoring. Basic Principles,
gies including nanowired drug delivery to Regeneration, Pathophysiology and Clinical
enhance neuroprotection. J Neural Transm Aspects, Eds: Stålberg E, Sharma H S, Olsson Y,
118, 155–176 Springer Wien New York, pp. 283–363
414 H.S. Sharma and A. Sharma
8. Fehlings MG, Tator CH (1988) A review of 18. Sharma HS (2007) Methods to produce
experimental models of acute spinal cord injury, hyperthermia-induced brain dysfunction. Prog
In: L Illis (Ed), Spinal Cord Dysfunction: Brain Res 162, 173–199
Assessment, Oxford University Press, Oxford, 19. Sharma HS (2005) Methods to produce brain
pp, 3–33 hyperthermia. Current Protocols in Toxicology
9. Sharma HS (2007) Neurotrophic factors in 23, UNIT 11.14, pp 1–26, DOI:
combination: a possible new therapeutic strat- 10.1002/0471140856.tx1114s23
egy to influence pathophysiology of spinal cord 20. Sharma HS (1999) Pathophysiology of
injury and repair mechanisms. Curr Pharm Des blood–brain barrier, brain edema and cell
13, 1841–1874 injury following hyperthermia: New role of
10. Sharma HS, Nyberg F, Gordh T, Alm P, heat shock protein, nitric oxide and carbon
Westman, J (1998) Neurotrophic factors atten- monoxide. An experimental study in the rat
uate neuronal nitric oxide synthase upregula- using light and electron microscopy, Acta
tion, microvascular permeability disturbances, Universitatis Upsaliensis 830, 1–94
edema formation and cell injury in the spinal 21. Sharma HS (2010) Selected combination of
cord following trauma. In: Spinal Cord neurotrophins potentiate neuroprotection and
Monitoring. Basic Principles, Regeneration, functional recovery following spinal cord injury
Pathophysiology and Clinical Aspects, Eds: in the rat. Acta Neurochir Suppl 106, 295–300
Stålberg E, Sharma HS, Olsson Y, Springer 22. Sharma HS (2007) A select combination of
Wien New York, pp. 181–210 neurotrophins enhances neuroprotection and
11. Sharma HS (2005) Pathophysiology of blood- functional recovery following spinal cord injury.
spinal cord barrier in traumatic injury and Ann NY Acad Sci 1122, 95–111
repair. Curr Pharm Des 11, 1353–1389 23. Sharma HS (2006) Post-traumatic application
12. Sharma HS (2003) Neurotrophic factors atten- of brain-derived neurotrophic factor and glia-
uate microvascular permeability disturbances derived neurotrophic factor on the rat spinal
and axonal injury following trauma to the rat cord enhances neuroprotection and improves
spinal cord. Acta Neurochir Suppl 86, motor function. Acta Neurochir Suppl 96,
383–388 329–334
13. Sharma HS, Olsson Y. (1990) Edema forma- 24. Sharma HS, Nyberg F, Gordh T, Alm P (2006)
tion and cellular alterations following spinal Topical application of dynorphin A (1–17)
cord injury in the rat and their modification antibodies attenuates neuronal nitric oxide
with p-chlorophenylalanine. Acta Neuropathol synthase up-regulation, edema formation, and
79, 604–610 cell injury following focal trauma to the rat
14. Olsson Y, Sharma HS, Pettersson CA (1990) spinal cord. Acta Neurochir 96, 309–315
Effects of p-chlorophenylalanine on microvas- 25. Sharma HS (2005) Neuroprotective effects of
cular permeability changes in spinal cord neurotrophins and melanocortins in spinal cord
trauma. An experimental study in the rat using injury: an experimental study in the rat using
131
I-sodium and lanthanum tracers. Acta pharmacological and morphological approaches.
Neuropathol 79, 595–603 Ann NY Acad Sci 1053, 407–421
15. Sharma HS (2009) Blood–Central Nervous 26. Sharma HS, Westman J, Gordh T, Alm P
System Barriers: The Gateway to (2000) Topical application of brain derived
Neurodegeneration, Neuroprotection and neurotrophic factor influences upregulation of
Neuroregeneration, In: A Lajtha, N Banik, SK constitutive isoform of heme oxygenase in the
Ray (Eds) Handbook of Neurochemistry and spinal cord following trauma an experimental
Molecular Neurobiology: Brain and Spinal study using immunohistochemistry in the rat.
Cord Trauma. Springer Verlag, Berlin, Acta Neurochir 76, 365–369
Heidelberg, New York, pp. 363–457 27. Winkler T, Sharma HS, Stålberg E, Badgaiyan
16. Winkler T, Sharma HS, Stålberg E, Olsson Y RD (2000) Neurotrophic factors attenuate
(1993) Indomethacin, an inhibitor of prosta- alterations in spinal cord evoked potentials and
glandin synthesis attenuates alteration in spinal edema formation following trauma to the rat
cord evoked potentials and edema formation spinal cord. Acta Neurochir 76, 291–296
after trauma to the spinal cord: an experimental 28. Sharma HS, Nyberg F, Gordh T, Alm P,
study in the rat. Neuroscience 52, 1057–1067 Westman J (2000) Neurotrophic factors influ-
17. Sharma HS, Winkler T, Stålberg E, Olsson Y, ence upregulation of constitutive isoform of
Dey PK (1991) Evaluation of traumatic spinal heme oxygenase and cellular stress response in
cord edema using evoked potentials recorded the spinal cord following trauma. An experi-
from the spinal epidural space. An experimental mental study using immunohistochemistry in
study in the rat. J Neurol Sci 102, 150–162 the rat. Amino Acids 19, 351–361
33 Rodent Spinal Cord Injury Model and Application of Neurotrophic… 415
29. Sharma HS, Nyberg F, Westman J, Alm P, rat spinal cord following trauma. Amino Acids
Gordh T, Lindholm D (1998) Brain-derived 23, 261–272
neurotrophic factor and insulin like growth 32. Allen AR (1911) Surgery of experimental lesion
factor-1 attenuate upregulation of nitric oxide of spinal cord equivalent to crush injury of
synthase and cell injury following trauma to the fracture dislocation of spinal column.
spinal cord. An immunohistochemical study in Preliminary report. JAMA 57, 878–880
the rat. Amino Acids 14, 121–129 33. Bresnahan CJ, Beattie MS, Todd FD, Noyes
30. Sharma HS, Nyberg F, Gordh T, Alm P, DH (1987) A behavioural and anatomical
Westman J (1997) Topical application of insu- analysis of spinal cord injury produced by a
lin like growth factor-1 reduces edema and feedback-controlled impaction device. Exp
upregulation of neuronal nitric oxide synthase Neurol 95, 548–570
following trauma to the rat spinal cord. Acta 34. Sharma HS, Dey PK (1982) Correlation of
Neurochir Suppl 70, 130–133 spinal cord tissue 5-HT with edema develop-
31. Sharma HS, Sjöquist P-O (2002) A new anti- ment following surgical spinal cord trauma in
oxidant compound H-290/51 modulates rats. Indian J Physiol Pharmacol 26, Supplement
glutamate and GABA immunoreactivity in the I, p. 8–10
INDEX
A 149–151, 153, 156, 161, 162, 165, 169, 172, 176, 177,
180–182, 184, 185, 191, 192, 194, 195, 216, 225, 227,
A2B5 monoclonal antibody ..............134, 139–142, 144, 245 230, 238, 239, 241, 243, 244, 250, 251, 297, 334–336,
Adeno-associated viral (AAV) vectors..................... 305–318 339, 340
Adrenal .................................................................... 223–234 Brain-derived neurotrophic factor (BDNF) ................... 3–6,
Adult....................... 14, 17, 19, 34, 39, 40, 67, 103–115, 117, 8, 24, 91–93, 96–98, 100, 104, 106, 108, 114, 117,
132, 138, 140–141, 143, 170, 171, 179–187, 120, 121, 125, 148, 150, 159, 161, 215, 216, 335,
189–199, 225, 230–232, 335, 384 338–340, 394, 395, 404, 406–411
Affinity purified antibodies ............................................. 139 BSA. See Bovine serum albumin (BSA)
Amperometric detection .................................................. 378 B27 supplement ............................. 25, 26, 41, 50, 51, 58, 59,
Amphotericin ................................... 191, 198, 345, 346, 352 62, 81, 82, 84–86, 92, 99, 104, 107, 108, 113, 119,
Amyloid β-peptide .................................8, 49, 57–64, 68, 85 121, 126, 133, 135, 143, 149, 151, 161, 163, 187, 297
Amyloid precursor protein (APP) ......57, 278, 289, 295–302
Anesthesia ....................................... 110, 358, 361, 364, 385, C
387–389, 398, 400, 401, 405, 410, 412 Campenot chambers ................................................ 214, 215
Antibody..................... 8, 17, 19, 45, 47, 93, 95, 97, 118, 122, Canine... ...........................................190–193, 195–196, 198
124, 127, 128, 134, 138–142, 149, 152–153, 192, Carbon nanotubes (CNTs) ...................................... 261–276
195, 197, 198, 235, 237–239, 241–246, 251, 253, Caspase .................................................................... 322, 323
254, 257, 308, 316, 317, 321–324, 327–330 Cell culture .............25, 26, 32–34, 40–41, 50, 53–55, 58–60,
Antigalactocerebroside antibody.............................. 134, 142 69, 72, 76, 80–85, 87, 92, 106, 117–128, 133, 134,
Apoptosis........................................ 23, 24, 31, 147, 213, 311 144, 149, 150, 161, 169, 181, 190, 202, 216, 218, 223,
APP. See Amyloid precursor protein (APP) 240, 248, 261–276, 279, 297, 307, 334, 343
Araldite............................................................ 395, 403, 407 Cell transduction ..................................................... 310–313
Ascorbic acid .....................357, 358, 361, 363, 366, 367, 378 Cerebellar granule neurons (CGNs) .................... 23–35, 160
Astrocytes ........... 44, 54, 67–76, 94, 117, 131, 132, 138, 139, Cerebellum ........ 23, 28, 29, 39–47, 52, 54, 71, 281, 282, 348
141–142, 144, 243–244, 266, 273, 308, 316 CGNs. See Cerebellar granule neurons (CGNs)
ATP................................................... 58, 59, 62–63, 210, 295 Chitosan .................................................................. 321–331
Automation ...............187, 249, 251, 254, 275, 276, 291, 315 Chloral hydrate ......................... 357, 359, 368, 374, 380, 395
Axonal transport .............................................. 214, 295–302 Chromaffin cells ...................................................... 223–234
Axons................................2, 13, 67, 117, 167–177, 179, 190, Ciliary neurotrophic factor (CNTF) .......................4, 8, 104,
202, 213, 247, 296, 357, 384 117, 120, 121, 134, 135, 141, 143, 148, 150, 152, 161,
163, 394–396
B
CNTs. See Carbon nanotubes (CNTs)
BBB. See Blood–brain barrier (BBB) Co-culture ................................................................... 79–88
BDNF. See Brain-derived neurotrophic factor (BDNF) Collagen ...........................114, 191, 198, 203, 204, 206, 209,
Biocompatible polymers .................................................. 322 214, 216–218, 220, 230, 232, 233, 251, 252, 258
Blood–brain barrier (BBB) ...................................... 321, 323 Collagenase ............................. 133, 135, 137–138, 141, 149,
Bone cutter ...................................................... 185, 396, 402 155, 161, 164, 171, 180, 181, 183, 185, 191, 194, 195,
Bovine......................... 26, 41, 44, 45, 61, 119, 133, 150, 161, 198, 225–227, 229, 230, 232, 233
191, 203, 224–230, 233, 250, 264, 273, 297, 309 Colorimetric assay ....................................................... 31, 58
Bovine serum albumin (BSA).................... 26, 27, 40, 42, 69, Compartmented chambers ...................................... 213–221
71, 97, 99, 100, 108, 114, 119, 120, 122, 123, Conditioned medium .......................104, 106–110, 114, 159
125–127, 133, 134, 136, 137, 139–142, 144, Cone photoreceptor cells ......................................... 147–157
Stephen D. Skaper (ed.), Neurotrophic Factors: Methods and Protocols, Methods in Molecular Biology, vol. 846,
DOI 10.1007/978-1-61779-536-7, © Springer Science+Business Media, LLC 2012
417
NEUROTROPHIC FACTORS: METHODS AND PROTOCOLS
418 Index
Confocal ..............45, 209, 220, 237, 245, 278–280, 284, 289 Fibroblast growth factor (FGF).........................4, 15, 24, 40,
Cortex.................................40, 49, 51–53, 67–76, 82, 85, 87, 41, 43, 91, 114, 117, 120, 121, 148, 190, 191, 193,
230, 244, 267, 268, 277, 283, 327, 330, 348 322–324, 326, 329, 330
Counterstain ..............................................45, 192, 196, 197, Fluorescence ............................................ 45, 47, 95, 96, 106,
242, 329, 407 168, 174, 192, 197, 209, 235–238, 244, 255, 265,
Coverslips..... ..................29, 45, 92, 132, 149, 160, 172, 181, 270, 275, 278, 280, 291, 314, 315, 317, 328, 329,
203, 214, 224, 240, 262, 284, 297, 306–307, 329 346, 347, 349–352
Crush... .....................................................383–386, 388, 389 Fluoromount-G....................................................... 239, 242
Cryoprotectant.... ............................................................ 107 Forskolin.................................. 120, 121, 134, 135, 141, 143,
Cyclic AMP.. ..............................................24, 104, 108, 143 150–152, 161–163, 191, 193
Cytospin .................................................................. 336, 337 Freezing ........................................................... 203, 205, 358
D G
Degeneration ......................................... 8, 88, 147, 148, 190, GDNF. See Glial cell line derived neurotrophic factor
202, 277, 344, 356, 357 (GDNF)
Degranulation.................................................................. 338 GFAP. See Glial fibrillary acidic protein (GFAP)
Dendrites ............................ 19, 179, 248, 277, 287, 291, 292 GFP. See Green fluorescent protein (GFP)
Dendritic spine ................................................ 277, 284–289 Glia.....................................67, 68, 74, 75, 79–88, 94–95, 99,
Deoxyribonuclease ..................................................... 40, 225 112, 131, 132, 147, 183, 184, 186, 221, 309, 310
Differential attachment ..................................................... 75 Glial cell line derived neurotrophic factor
3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (GDNF) .............................. 4, 8, 104, 106, 108, 114,
bromide (MTT) ...................... 16, 25–27, 31–33, 35, 120, 148, 159, 394, 395, 404, 406, 408, 410, 411
81, 86, 88, 96, 120, 125, 127 Glial fibrillary acidic protein (GFAP) ...................44, 73, 94,
Dinitrophenol (DNP) antibody....................... 335, 337, 338 239, 244, 245, 308, 311
Direct immunofluorescence ..................................... 237, 238 Glutamate...................................... 15, 16, 34, 67, 68, 93, 94,
Dispase .................................................................... 191, 194 126, 307, 314, 315, 317, 343, 346, 349–352
Dissection ................................ 25, 28–29, 32, 40–43, 45, 46, Glutaraldehyde ................................................ 335, 337, 395
50–54, 68, 70–71, 74, 92, 104, 118, 120–122, Glycogen synthase kinase-3 (GSK-3) ............................... 24
125, 132, 148, 160, 182–185, 187, 203, 206, Gradient ............. 53, 104, 105, 108, 111–113, 168, 170, 171,
207, 230, 262, 265–269, 274, 279–283, 289, 173–177, 219, 224, 229, 234, 336, 340, 366, 373, 376
290, 344, 385, 388 Green fluorescent protein (GFP) ....................209, 278–280,
Dissociation ....................27–29, 35, 52, 54, 74, 99, 111, 112, 307, 311, 313–317
120–122, 125, 126, 138, 139, 153–156, 164, 180, Growth cones .................................. 13, 14, 17, 19, 167–177,
181, 183–186, 190, 191, 193–194, 230, 231, 233 190, 217, 220, 248, 270, 271, 276
Dopamine ...............................91, 92, 98, 159, 201, 205, 210, GSK-3. See Glycogen synthase kinase-3 (GSK-3) ............ 24
355, 363, 365, 367
Dorsal root ganglion (DRG) ...........................168–172, 174, H
176, 177, 179–187, 190, 215, 216, 218–219, 296
Hamilton syringe .....................................357, 358, 361–363,
E 370, 373, 376, 377, 396
Hemocytometer....................25, 29–30, 35, 50, 82, 118, 132,
Embryonic ................................34, 40, 49, 51–55, 58, 60, 80, 149, 160, 169, 172, 184, 186, 187, 224, 226, 233, 252
87, 92, 103, 120, 125, 132, 138, 142, 176, 180, 318 High content analysis .............................................. 247–259
Enrichment ..............................................190, 192, 194–196 High performance liquid chromatography
Eppendorf tube ................................. 30, 100, 118, 132, 137, (HPLC) ........................ 366, 368, 370, 373, 377–378
172, 244, 327, 340, 346, 352, 353, 358 Hippocampus .................................. 39, 49, 51–55, 265, 268,
Evans blue ........................................395, 404–407, 410, 412 277, 280–284, 343, 348, 353
Excitotoxicity................................ 15, 68, 311, 343, 345, 349 Horse serum .................................... 107, 203, 205–207, 209,
215, 216, 220, 279, 289, 345, 353
F
Human ...................................3, 14, 39, 59, 67, 91, 119, 133,
FGI. See Fibroblast growth factor (FGF) 147, 161, 180, 189–194, 196–198, 202, 216, 248,
Fiber optic cold light source ............................................ 347 329, 333, 355, 366, 393
Fibroblast ........................................................141, 189, 190, 6-Hydroxydopamine (6-OHDA) ....................159, 202, 204,
192, 194–199, 234 208, 210, 355–364
NEUROTROPHIC FACTORS: METHODS AND PROTOCOLS
Index
419
Oligodendrocytes .............................. 14, 15, 44, 67–76, 117, Rat........ ................................... 19, 23–35, 39–47, 49, 51–55,
131, 132, 135, 137, 139, 141, 142 57–64, 70–71, 80, 82, 85, 87, 91–100, 103,
Optic nerve ..........52, 117, 118, 121, 127, 131–144, 155, 164 118, 121, 124–125, 131, 147–157, 159–166,
Osmium tetroxide.................................................... 395, 407 168, 179–187, 189–199, 201–210, 216, 223,
Ovomucoid .................................27, 28, 52, 53, 71, 119, 120, 244, 262, 313, 329, 333–340, 343–353, 356,
122, 126, 133, 134, 136–138, 144, 149–151, 365, 383–390, 394
154–156, 161, 162, 164, 165 Release assay ................................................ 60–62, 333–340
Oxygen-glucose deprivation (OGD) ...................... 344–346, Retina.. ............................................ 118, 121–123, 125, 126,
349, 351–353 147, 148, 153–155, 159–166
Retinal ganglion cells (RGCs) ................................. 117–128
P RGCs. See Retinal ganglion cells (RGCs)
Paddle pastettes ............................................... 347, 348, 353
S
Papain.. .............................26–29, 50, 52–54, 69, 71, 74, 107,
108, 113, 118–120, 122, 125–127, 133, 135–139, SCG. See Superior cervical ganglia (SCG)
141, 142, 144, 149, 150, 153, 155, 161, 162, 164, Schwann cells ..................................................118, 189–199,
225, 232, 263, 264, 266, 268, 273, 297, 298, 302 221, 384
Paraformaldehyde .................................... 41, 44, 45, 99, 192, Sciatic nerve .................................................... 296, 383–390
197, 238, 244, 251, 253, 279, 395, 407 Selenite ....................................................119–121, 133, 135,
Parkinson’s disease models ....................... 201–210, 355–364 150, 151, 161–163, 169
PC12. See Pheochromocytoma cells (PC12) Sensory neurons ...................................1, 179–187, 213, 214
Peanut lectin .................................................................... 148 Serotonin ................................................................. 333–340
PEG. See Polyethylene glycol (PEG) Slice cultures ............................................180, 262, 277–292,
PEI. See Polyethyleneimine (PEI) 343–353
Peptide delivery ....................................................... 321–331 Spinal cord....................................... 17, 40, 67–76, 103–115,
Peristaltic pump ............................................... 385, 388, 405 160, 170, 179, 215, 267, 384, 393
Peritoneal fluid ................................................................ 336 Spinal cord injury .........................................8, 104, 393–413
Phase contrast optics ........................ 32, 86, 96, 97, 337, 353 Sprague–Dawley .......................................121, 262, 266, 273
Pheochromocytoma cells (PC12) ........................... 201–210, Stereo microscope....................................191, 198, 225, 232,
248–250, 258 347, 348, 370, 396, 402
Phosphatidylinositol 3-kinase (PI 3-kinase).........6, 7, 24, 25 Stereotaxic surgery........................................... 359, 369–370
Photoreceptor cells .................................................. 147–157 Sterilin plastic dishes ..................................68, 72, 73, 75, 80
PI. See Propidium iodide (PI) Streptavidin .............................................238, 246, 308, 316,
Pigment epithelium ......................................... 148, 159–166 322, 324, 325, 327
Platelet-derived growth factor (PDGF) ..................... 3, 134, Substance P ..................................................... 335, 338, 339
140, 143, 148, 159 Substratum .................................. 27, 70, 73, 81–82, 94, 169,
Polyethylene glycol (PEG) ..............................262, 263, 270, 182, 216–218, 220, 221
271, 322–327 Superior cervical ganglia (SCG) ............................. 201–210,
Polyethyleneimine (PEI) ........................................ 262, 265, 214, 216, 218–219
266, 268, 270–272, 274 Sympathetic neurons ............................8, 201–210, 214, 220
Poly-L-lysine ....................................... 26, 27, 31, 34, 69, 70,
72–74, 94–96, 99, 169, 181, 186, 191, 238, 240, 244, T
306–308, 313, 317 T3. See 3,3’,5-Triiodo-L-thyronine (T3) .................. 69, 119,
Poly-L-ornithine .................... 34, 95, 99, 181, 182, 191, 233 133, 150, 161
Progenitors ................... 39–47, 131, 132, 137–144, 148, 160 Targeted brain delivery ............................................ 321–331
Progesterone ............................................119–121, 133, 135, Thawing, cell lines ................................................... 204–205
150, 151, 161–163, 169 Thyroxine .......................................................... 69, 119, 133
Propidium iodide (PI) ..................................... 346, 349–353 Toluidine blue...........................................349, 395, 407, 408
Putrescine ................................................119, 120, 133, 135, Transection ....................................... 384, 386, 387, 390, 398
150, 151, 161, 162, 169 Transfection ..................................... 168, 169, 172, 174, 176,
177, 202, 209, 297–299, 302, 305
R
Transferrin ................................. 70, 119, 120, 133, 134, 150,
RAN-2 antibody .............................................120, 127, 134, 151, 161, 162, 169, 321–324
138, 139, 141, 144 3,3’,5-Triiodo-L-thyronine (T3) ........ 69, 119, 133, 150, 161
NEUROTROPHIC FACTORS: METHODS AND PROTOCOLS
Index
421
Trypan blue ....................................... 26, 30, 35, 43, 44, 112, V
120, 124, 134, 191, 194–196, 218, 224, 226, 258,
307, 309, 334, 336 VectaShield ............................... 239, 242, 245, 308, 314, 316
Trypsin .......................................... 26, 40, 68, 107, 119, 133, Viral vector .................................................................. 8, 305
149, 161, 169, 180, 192, 203, 251, 264, 307
W
Tyrosine hydroxylase.................................................... 95, 97
Western blot .................................................................... 312
U
Y
Uranyl acetate .......................................................... 395, 407
Uric acid .......................................................... 366, 367, 378 Yellow fluorescent protein (YFP) ............................ 297–300