Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Protocol
  • Published:

Targeted intraspinal injections to assess therapies in rodent models of neurological disorders

Abstract

Despite decades of research, pharmacological therapies for spinal cord motor pathologies are limited. Alternatives using macromolecular, viral, or cell-based therapies show early promise. However, introducing these substances into the spinal cord, past the blood–brain barrier, without causing injury is challenging. We describe a technique for intraspinal injection targeting the lumbar ventral horn in rodents. This technique preserves motor performance and has a proven track record of translation into phase 1 and 2 clinical trials in amyotrophic lateral sclerosis (ALS) patients. The procedure, in brief, involves exposure of the thoracolumbar spine and dissection of paraspinous muscles over the target vertebrae. Following laminectomy, the spine is affixed to a stereotactic frame, permitting precise and reproducible injection throughout the lumbar spine. We have used this protocol to inject various stem cell types, primarily human spinal stem cells (HSSCs); however, the injection is adaptable to any candidate therapeutic cell, virus, or macromolecule product. In addition to a detailed procedure, we provide stereotactic coordinates that assist in targeting of the lumbar spine and instructional videos. The protocol takes ~2 h per animal.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Schematics depicting laminectomy for exposure of rodent lumbar spinal cord and cross-sectional views depicting coordinates for optimal targeting of the ventral horn.
Fig. 2: Intraspinal injection of stem cells into SOD1G93A mouse lumbar spine.
Fig. 3: Intraspinal injection of stem cells into SOD1G93A rat lumbar spine.
Fig. 4: Intraspinal injection of stem cells does not impair motor performance in SOD1G93A mice or rats.
Fig. 5: Successful targeting of stem cell injections to the ventral horn of the lumbar spinal cord of the mutant SOD1G93A mouse.

Similar content being viewed by others

Data availability

The data presented with the protocol are available from the corresponding author on reasonable request.

References

  1. Ashizawa, T., Oz, G. & Paulson, H. L. Spinocerebellar ataxias: prospects and challenges for therapy development. Nat. Rev. Neurol. 14, 590–605 (2018).

    Article  Google Scholar 

  2. Groen, E. J. N., Talbot, K. & Gillingwater, T. H. Advances in therapy for spinal muscular atrophy: promises and challenges. Nat. Rev. Neurol. 14, 214–224 (2018).

    Article  Google Scholar 

  3. Hardiman, O. et al. Amyotrophic lateral sclerosis. Nat. Rev. Dis. Primers 3, 17071 (2017).

    Article  Google Scholar 

  4. Teoh, H. L. et al. Inherited paediatric motor neuron disorders: beyond spinal muscular atrophy. Neural Plast. 2017, 6509493 (2017).

    Article  Google Scholar 

  5. Ahuja, C. S. et al. Traumatic spinal cord injury. Nat. Rev. Dis. Primers 3, 17018 (2017).

    Article  Google Scholar 

  6. Langhorne, P., Coupar, F. & Pollock, A. Motor recovery after stroke: a systematic review. Lancet Neurol. 8, 741–754 (2009).

    Article  Google Scholar 

  7. Gubert, F. et al. Intraspinal bone-marrow cell therapy at pre- and symptomatic phases in a mouse model of amyotrophic lateral sclerosis. Stem Cell Res. Ther. 7, 41 (2016).

    Article  Google Scholar 

  8. Inquimbert, P., Moll, M., Kohno, T. & Scholz, J. Stereotaxic injection of a viral vector for conditional gene manipulation in the mouse spinal cord. J. Vis. Exp. 2013, e50313 (2013).

  9. Knippenberg, S. et al. Intraspinal administration of human spinal cord-derived neural progenitor cells in the G93A-SOD1 mouse model of ALS delays symptom progression, prolongs survival and increases expression of endogenous neurotrophic factors. J. Tissue Eng. Regen. Med. 11, 751–764 (2017).

    Article  CAS  Google Scholar 

  10. Kohro, Y. et al. A new minimally-invasive method for microinjection into the mouse spinal dorsal horn. Sci. Rep. 5, 14306 (2015).

    Article  CAS  Google Scholar 

  11. Lepore, A. C. Intraspinal cell transplantation for targeting cervical ventral horn in amyotrophic lateral sclerosis and traumatic spinal cord injury. J. Vis. Exp. 2011, 3069 (2011).

    Google Scholar 

  12. Hefferan, M. P. et al. Human neural stem cell replacement therapy for amyotrophic lateral sclerosis by spinal transplantation. PLoS ONE 7, e42614 (2012).

    Article  CAS  Google Scholar 

  13. Xu, L., Ryugo, D. K., Pongstaporn, T., Johe, K. & Koliatsos, V. E. Human neural stem cell grafts in the spinal cord of SOD1 transgenic rats: differentiation and structural integration into the segmental motor circuitry. J. Comp. Neurol. 514, 297–309 (2009).

    Article  CAS  Google Scholar 

  14. Xu, L., Shen, P., Hazel, T., Johe, K. & Koliatsos, V. E. Dual transplantation of human neural stem cells into cervical and lumbar cord ameliorates motor neuron disease in SOD1 transgenic rats. Neurosci. Lett. 494, 222–226 (2011).

    Article  CAS  Google Scholar 

  15. Xu, L. et al. Human neural stem cell grafts ameliorate motor neuron disease in SOD-1 transgenic rats. Transplantation 82, 865–875 (2006).

    Article  Google Scholar 

  16. Yan, J. et al. Combined immunosuppressive agents or CD4 antibodies prolong survival of human neural stem cell grafts and improve disease outcomes in amyotrophic lateral sclerosis transgenic mice. Stem Cells 24, 1976–1985 (2006).

    Article  CAS  Google Scholar 

  17. Yan, J. et al. Extensive neuronal differentiation of human neural stem cell grafts in adult rat spinal cord. PLoS Med. 4, e39 (2007).

    Article  Google Scholar 

  18. Goutman, S. A. et al. Long-term phase 1/2 intraspinal stem cell transplantation outcomes in ALS. Ann. Clin. Transl. Neurol. 5, 730–740 (2018).

  19. Feldman, E. L. et al. Intraspinal neural stem cell transplantation in amyotrophic lateral sclerosis: Phase 1 trial outcomes. Ann. Neurol. 75, 363–373 (2014).

    Article  Google Scholar 

  20. Glass, J. D. et al. Lumbar intraspinal injection of neural stem cells in patients with amyotrophic lateral sclerosis: results of a phase I trial in 12 patients. Stem Cells 30, 1144–1151 (2012).

    Article  CAS  Google Scholar 

  21. Glass, J. D. et al. Transplantation of spinal cord-derived neural stem cells for ALS: analysis of phase 1 and 2 trials. Neurology 87, 392–400 (2016).

    Article  CAS  Google Scholar 

  22. Riley, J. et al. Intraspinal stem cell transplantation in amyotrophic lateral sclerosis: a phase I safety trial, technical note, and lumbar safety outcomes. Neurosurgery 71, 405–416 (2012). discussion 416.

    Article  Google Scholar 

  23. Riley, J. et al. Intraspinal stem cell transplantation in amyotrophic lateral sclerosis: a phase I trial, cervical microinjection, and final surgical safety outcomes. Neurosurgery 74, 77–87 (2014).

    Article  Google Scholar 

  24. Hardcastle, N., Boulis, N. M. & Federici, T. AAV gene delivery to the spinal cord: serotypes, methods, candidate diseases, and clinical trials. Expert Opin. Biol. Ther. 18, 293–307 (2018).

    Article  CAS  Google Scholar 

  25. Schoch, K. M. & Miller, T. M. Antisense oligonucleotides: translation from mouse models to human neurodegenerative diseases. Neuron 94, 1056–1070 (2017).

    Article  CAS  Google Scholar 

  26. Cizkova, D. et al. Functional recovery in rats with ischemic paraplegia after spinal grafting of human spinal stem cells. Neuroscience 147, 546–560 (2007).

    Article  CAS  Google Scholar 

  27. Gordon, P. H. Amyotrophic lateral sclerosis: an update for 2013 clinical features, pathophysiology, management and therapeutic trials. Aging Dis. 4, 295–310 (2013).

    Article  Google Scholar 

  28. Bensimon, G., Lacomblez, L. & Meininger, V. A controlled trial of riluzole in amyotrophic lateral sclerosis. ALS/Riluzole Study Group. N. Engl. J. Med. 330, 585–591 (1994).

    Article  CAS  Google Scholar 

  29. Writing Group & Edaravone (MCI-186) ALS 19 Study Group. Safety and efficacy of edaravone in well defined patients with amyotrophic lateral sclerosis: a randomised, double-blind, placebo-controlled trial. Lancet Neurol. 16, 505–512 (2017).

  30. Chen, K. S., Sakowski, S. A. & Feldman, E. L. Intraspinal stem cell transplantation for amyotrophic lateral sclerosis. Ann. Neurol. 79, 342–353 (2016).

    Article  Google Scholar 

  31. Lunn, J. S., Sakowski, S. A. & Feldman, E. L. Stem cell therapies for amyotrophic lateral sclerosis: Recent advances and prospects for the future. Stem Cells 32, 1099–1109 (2014).

    Article  CAS  Google Scholar 

  32. Sakowski, S. A., Lunn, J. S. & Feldman, E. L. Stem cell therapy for motor neuron disease. in Motor Neuron Disease in Adults, Contemporary Neurology Series, Vol. 88 (eds Bruijn, L. & Bromberg, M. B.) 312–317 (Oxford University Press, New York, 2015).

  33. Ilieva, H., Polymenidou, M. & Cleveland, D. W. Non-cell autonomous toxicity in neurodegenerative disorders: ALS and beyond. J. Cell Biol. 187, 761–772 (2009).

    Article  CAS  Google Scholar 

  34. Paez-Colasante, X., Figueroa-Romero, C., Sakowski, S. A., Goutman, S. A. & Feldman, E. L. Amyotrophic lateral sclerosis: mechanisms and therapeutics in the epigenomic era. Nat. Rev. Neurol. 11, 266–279 (2015).

    Article  CAS  Google Scholar 

  35. Boulis, N. M. et al. Translational stem cell therapy for amyotrophic lateral sclerosis. Nat. Rev. Neurol. 8, 172–176 (2011).

    Article  Google Scholar 

  36. Goutman, S. A., Chen, K. S. & Feldman, E. L. Recent advances and the future of stem cell therapies in amyotrophic lateral sclerosis. Neurotherapeutics 12, 428–448 (2015).

    Article  CAS  Google Scholar 

  37. Thomsen, G. M., Gowing, G., Svendsen, S. & Svendsen, C. N. The past, present and future of stem cell clinical trials for ALS. Exp. Neurol. 262, 127–137 (2014).

    Article  CAS  Google Scholar 

  38. Johe, K. K., Hazel, T. G., Muller, T., Dugich-Djordjevic, M. M. & McKay, R. D. Single factors direct the differentiation of stem cells from the fetal and adult central nervous system. Genes Dev. 10, 3129–3140 (1996).

    Article  CAS  Google Scholar 

  39. Raore, B. et al. Cervical multilevel intraspinal stem cell therapy: assessment of surgical risks in Gottingen minipigs. Spine 36, E164–E171 (2011).

    Article  Google Scholar 

  40. Riley, J. et al. Targeted spinal cord therapeutics delivery: stabilized platform and MER guidance validation. Stereotact. Funct. Neurosurg. 86, 67–74 (2007).

    Article  Google Scholar 

  41. Riley, J. et al. Cervical spinal cord therapeutics delivery: preclinical safety validation of a stabilized microinjection platform. Neurosurgery 65, 754–761 (2009). discussion 761-762.

    Article  Google Scholar 

  42. Riley, J. P., Raore, B., Taub, J. S., Federici, T. & Boulis, N. M. Platform and cannula design improvements for spinal cord therapeutics delivery. Neurosurgery 69, 147–155 (2011).

    Google Scholar 

  43. Usvald, D. et al. Analysis of dosing regimen and reproducibility of intraspinal grafting of human spinal stem cells in immunosuppressed minipigs. Cell Transplant. 19, 1103–1122 (2010).

    Article  Google Scholar 

  44. Hefferan, M. P. et al. Optimization of immunosuppressive therapy for spinal grafting of human spinal stem cells in a rat model of ALS. Cell Transplant. 20, 1153–1161 (2011).

    Article  Google Scholar 

  45. McGoldrick, P., Joyce, P. I., Fisher, E. M. & Greensmith, L. Rodent models of amyotrophic lateral sclerosis. Biochim. Biophys. Acta 1832, 1421–1436 (2013).

    Article  CAS  Google Scholar 

  46. Philips, T. & Rothstein, J. D. Rodent models of amyotrophic lateral sclerosis. Curr. Protoc. Pharmacol. 69, 5.67.1–5.67.21 (2015).

    Article  Google Scholar 

  47. Xu, L., Shen, P., Hazel, T., Johe, K. & Koliatsos, V. E. Dual transplantation of human neural stem cells into cervical and lumbar cord ameliorates motor neuron disease in SOD1 transgenic rats. Neurosci. Lett. 494, 222–226 (2011).

    Article  CAS  Google Scholar 

  48. Franz, C. K. et al. Intraspinal cord delivery of IGF-I mediated by adeno-associated virus 2 is neuroprotective in a rat model of familial ALS. Neurobiol. Dis. 33, 473–481 (2009).

    Article  CAS  Google Scholar 

  49. Lepore, A. C. et al. Intraparenchymal spinal cord delivery of adeno-associated virus IGF-1 is protective in the SOD1G93A model of ALS. Brain Res. 1185, 256–265 (2007).

    Article  CAS  Google Scholar 

  50. Donnelly, E. M. et al. Lentiviral vector delivery of short hairpin RNA to NG2 and neurotrophin-3 promotes locomotor recovery in injured rat spinal cord. Cytotherapy 14, 1235–1244 (2012).

    Article  CAS  Google Scholar 

  51. Macks, C., Gwak, S. J., Lynn, M. & Lee, J. S. Rolipram-loaded polymeric micelle nanoparticle reduces secondary injury after rat compression spinal cord injury. J. Neurotrauma 35, 582–592 (2018).

    Article  Google Scholar 

  52. Povysheva, T. et al. Post-spinal cord injury astrocyte-mediated functional recovery in rats after intraspinal injection of the recombinant adenoviral vectors Ad5-VEGF and Ad5-ANG. J. Neurosurg. Spine 27, 105–115 (2017).

    Article  Google Scholar 

  53. Santamaria, A. J., Solano, J. P., Benavides, F. D. & Guest, J. D. Intraspinal delivery of Schwann cells for spinal cord injury. Methods Mol. Biol. 1739, 467–484 (2018).

    Article  CAS  Google Scholar 

  54. Brown, R. H. Jr. & Al-Chalabi, A. Amyotrophic lateral sclerosis. N. Engl. J. Med. 377, 162–172 (2017).

    Article  CAS  Google Scholar 

  55. Parente, V. & Corti, S. Advances in spinal muscular atrophy therapeutics. Ther. Adv. Neurol. Disord. 11, 1756285618754501 (2018).

    Article  Google Scholar 

  56. Scoles, D. R. et al. Antisense oligonucleotide therapy for spinocerebellar ataxia type 2. Nature 544, 362–366 (2017).

    Article  CAS  Google Scholar 

  57. Thuret, S., Moon, L. D. & Gage, F. H. Therapeutic interventions after spinal cord injury. Nat. Rev. Neurosci. 7, 628–643 (2006).

    Article  CAS  Google Scholar 

  58. Garbuzova-Davis, S. et al. Positive effect of transplantation of hNT neurons (NTera 2/D1 cell-line) in a model of familial amyotrophic lateral sclerosis. Exp. Neurol. 174, 169–180 (2002).

    Article  Google Scholar 

  59. Bell, J. A., Sharpe, L. G. & Berry, J. N. Depressant and excitant effects of intraspinal microinjections of morphine and methionine-enkephalin in the cat. Brain Res. 196, 455–465 (1980).

    Article  CAS  Google Scholar 

  60. Garbuzova-Davis, S. et al. Intraspinal implantation of hNT neurons into SOD1 mice with apparent motor deficit. Amyotroph. Lateral Scler. Other Motor Neuron Disord. 2, 175–180 (2001).

    Article  CAS  Google Scholar 

  61. Qiu, K., Falk, D. J., Reier, P. J., Byrne, B. J. & Fuller, D. D. Spinal delivery of AAV vector restores enzyme activity and increases ventilation in Pompe mice. Mol. Ther. 20, 21–27 (2012).

    Article  CAS  Google Scholar 

  62. Dutta, S. & Sengupta, P. Men and mice: relating their ages. Life Sci. 152, 244–248 (2016).

    Article  CAS  Google Scholar 

  63. Sengupta, P. The laboratory rat: relating its age with human’s. Int. J. Prev. Med. 4, 624–630 (2013).

    PubMed  PubMed Central  Google Scholar 

  64. Lunn, J. S. et al. Intraspinal transplantation of neurogenin-expressing stem cells generates spinal cord neural progenitors. Neurobiol. Dis. 46, 59–68 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank C. Backus, C. Pacut, and M. Sweeney for technical support. This work was supported by the Sinai Medical Foundation, the A. Alfred Taubman Medical Research Institute, the Program for Neurology Research & Discovery, University of Michigan Clinician Scientist Training Program grants NINDS R25NS089450 (K.S.C.) and NIH T32NS07222 (O.N.K.), and the Robert E. Nederlander Sr. Program for Alzheimer’s Research (L.M.M.).

Author information

Authors and Affiliations

Authors

Contributions

E.L.F., K.S.C., L.M.M., O.N.K, and K.J. conceived of and designed the experiments. K.S.C. and O.N.K. performed the surgery with J.M.H., E.S.B., and J.S.C. F.E.M. J.S.C., and M.A.T. performed behavioral testing. L.M.M., E.S.B., and J.M.H. performed tissue processing, histological analyses, and microscopy. K.S.C. and L.M.M. analyzed and interpreted the data, and E.L.F. supervised the study. K.S.C., L.M.M., and S.A.S wrote the manuscript, and all authors reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Eva L. Feldman.

Ethics declarations

Competing interests

K.J. is chief scientific officer for Neuralstem, which provided the stem cell product (NSI566-RSC) via a material transfer agreement. The other authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

Key references using this protocol

Hefferan, M. P. et al. PLoS ONE 7, e42614 (2012): https://doi.org/10.1371/journal.pone.0042614

Yan, J. et al. Stem Cells 24, 1976–1985 (2006): https://doi.org/10.1634/stemcells.2005-0518

Lunn, J. S. et al. Neurobiol. Dis. 46, 59–68 (2012): https://doi.org/10.1016/j.nbd.2011.12.044

Integrated supplementary information

Supplementary Figure 1 Effect of intraspinal injection on animal survival and average number of surviving motor neurons.

Kaplan-Meier survival and quantification of postmortem motor neuron (MN) survival in wild type mice (a,b) and rats (c,d) undergoing vehicle intraspinal injection (WT Veh) versus animals undergoing no surgical procedure (WT). Motor neuron survival was assessed as described in the Supplementary Methods. Non-significant hazard ratio of mortality for animals undergoing intraspinal injection; minimal mortality (10% mortality for both mice (a) and rats (c) in the WT Veh animals could also be attributable to immunosuppressive regimen, as these animals served as controls in other stem cell experiments requiring immunosuppression. Likewise, quantified average number of surviving MN (mean ± SD) are not impacted by the injection protocol at approximately 180 days post-surgery, in mice (b; p=0.2762; WT vs. WT Veh; t-test) or in rats (d; p=0.1445; WT vs. WT Veh; t-test). MN = motor neuron; WT = wild-type; WT Veh = WT plus vehicle injection.

Supplementary information

Supplementary Text and Figures

Supplementary Figure 1 and Supplementary Methods

Reporting Summary

Supplementary Video 1

Technique for intraspinal injection of stem cells in SOD1G93A mouse lumbar spine. An incision is made over the lumbar area, and paraspinous muscles are sharply dissected free from spinous processes and spinal laminae. Cutting the ligamentous attachment to the facet joint facilitates exposure. By palpating the final rib, a laminectomy is performed at the T13 and L1 level using microscissors. The central spinal vein is used as the reference for midline. The needle tip is introduced 0.3 mm lateral to the 29 central vein and 0.8 mm deep to the dorsal surface of the spinal cord to reach the ventral gray matter. Five bilateral injections (10 total injections) are made with a 1.0-mm distance between each set of injections. All animal experiments were approved by the University of Michigan Institutional Animal Care and Use Committee and were performed in accordance with University of Michigan guidelines (accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care International) and state and federal regulations.

Supplementary Video 2

Technique for intraspinal injection of stem cells in SOD1G93A rat lumbar spine. An incision is made over the lumbar area, and paraspinous muscles are sharply dissected free from spinous processes and spinal laminae. Cutting the ligamentous attachment to the facet joint facilitates exposure. By palpating the final rib, a laminectomy is performed at the T13 level using a diamond-tip drill. The central spinal vein is used as the reference for midline. The needle tip is introduced 0.5 mm lateral to the central vein and 1.5 mm deep to the dorsal surface of the spinal cord to reach the ventral gray matter. Ten bilateral injections (20 total injections) are made with 0.5-mm distance between each set of injections. Fasciae of the subcutaneous muscle and skin are closed with absorbable sutures. All animal experiments were approved by the University of Michigan Institutional Animal Care and Use Committee and were performed in accordance with University of Michigan guidelines (accredited by the Association for the Assessment and Accreditation of Laboratory Animal Care International) and state and federal regulations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, K.S., McGinley, L.M., Kashlan, O.N. et al. Targeted intraspinal injections to assess therapies in rodent models of neurological disorders. Nat Protoc 14, 331–349 (2019). https://doi.org/10.1038/s41596-018-0095-5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-018-0095-5

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research