Using a Transection Paradigm to Enhance the Repair Mechanisms of an Investigational Human Cell Therapy

Monica J Chau, Jorge E Quintero, Paula V Monje, Stephen Randal Voss, Andrew S Welleford, Greg A Gerhardt, Craig G van Horne, Monica J Chau, Jorge E Quintero, Paula V Monje, Stephen Randal Voss, Andrew S Welleford, Greg A Gerhardt, Craig G van Horne

Abstract

One promising strategy in cell therapies for Parkinson's disease (PD) is to harness a patient's own cells to provide neuroprotection in areas of the brain affected by neurodegeneration. No treatment exists to replace cells in the brain. Thus, our goal has been to support sick neurons and slow neurodegeneration by transplanting living repair tissue from the peripheral nervous system into the substantia nigra of those with PD. Our group has pioneered the transplantation of transection-activated sural nerve fascicles into the brain of human subjects with PD. Our experience in sural nerve transplantation has supported the safety and feasibility of this approach. As part of a paradigm to assess the reparative properties of human sural nerve following a transection injury, we collected nerve tissue approximately 2 weeks after sural nerve transection for immunoassays from 15 participants, and collected samples from two additional participants for single nuclei RNA sequencing. We quantified the expression of key neuroprotective and select anti-apoptotic genes along with their corresponding protein levels using immunoassays. The single nuclei data clustered into 10 distinctive groups defined on the basis of previously published cell type-specific genes. Transection-induced reparative peripheral nerve tissue showed RNA expression of neuroprotective factors and anti-apoptotic factors across multiple cell types after nerve injury induction. Key proteins of interest (BDNF, GDNF, beta-NGF, PDGFB, and VEGF) were upregulated in reparative tissue. These results provide insight on this repair tissue's utility as a neuroprotective cell therapy.

Keywords: cell therapy; neuroprotection; peripheral nerve; single nuclei RNA sequencing; tissue-based therapy.

Conflict of interest statement

Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Figures

Figure 1.
Figure 1.
Study overview. This overview illustrates our sural nerve transection approach and subsequent tissue collection of the naïve and reparative nerve tissues. One to two centimeters of nerve was excised, which we called “naïve” nerve tissue. Approximately 14 days after, 1 to 2 centimeters from the distal nerve stump of the same nerve was excised, which we called “reparative” nerve tissue. Cross-sections of reparative sural nerve were stained with H&E (left) and Luxol fast blue (LBF)/MCOLL staining to show myelin and collagen. Individual nerve fascicles were separated, snap-frozen, and used for single nuclei RNA sequencing and immunoassays or implanted directly into the brain as part of our clinical trial. Shown here is a 3D view of reparative nerve fascicles implanted into the substantia nigra in the cell therapy trial. H&E: hematoxylin and eosin; CNS: central nervous system; 3D: three dimensional.
Figure 2.
Figure 2.
Reparative peripheral nerve tissue contains several cell types including regenerating cells. (A) Aggregate of single nuclei RNA sequencing from two participants. Ten unique cell type clusters were present in the reparative tissue. (B) Data from two participants show similarity in their cellular profiles. (C) The data clustered into 10 cell groups and were defined based on characteristic genes previously published for each particular cell type. (D) Each cell cluster’s percentage of total number of cells is reported.
Figure 3.
Figure 3.
Reparative peripheral nerve tissue shows RNA expression of neuroprotective factors 2 weeks after nerve transection. (A) Single nuclei RNA sequencing UMAP plots show cell-type expression of the neuroprotective factors of interest: NGF, PDGFB, VEGFA, PDGFA, CDNF, GDNF, BDNF, and EPO. The accompanying violin plots show the relative RNA expression level (log2 average) and frequencies for each cell type in the reparative peripheral nerve tissue. NGF, PDGFA, PDGFB, and VEGF were localized to more than one cell type. NGF was localized to the heterogeneous cell cluster and endoneurial mesenchymal cells, NGFR was localized to Schwann cells and the heterogeneous cell cluster, PDGFA was localized to myelinating Schwann cells and pericytes/vascular smooth muscle cells, PDGFB was highly expressed in endothelial cells. Cell type key: SC: Schwann cell; mySC: myelinating Schwann cells; EC: endothelial cells; PC: proliferating cells; HC: heterogeneous cells; EMC: endoneurial mesenchymal cells; MC: mesenchymal cells; M: macrophages; PC/VMSC: pericytes/vascular smooth muscle cells; TC: T-cells. (B) Repair Schwann cells are a source of neuroprotective factor release. NGFR is an abundantly expressed repair Schwann cell marker that was found in the Schwann and heterogeneous cell clusters. (C) Percentage of total cells expressing each key neuroprotective factors. Note that the x-axis maximum is 20%. Data obtained from N = 2 participants. BDNF: brain-derived neurotrophic factor; CDNF: cerebral dopamine neurotrophic factor; GDNF: glial-cell derived neurotrophic factor; NGF: nerve growth factor; NGFR: nerve growth factor receptor; PDGFA: platelet-derived growth factor–A; PDGFB: platelet-derived growth factor–B; VEGFA: vascular endothelial growth factor A; EPO: erythropoietin; UMAP: Uniform Manifold Approximation and Projection for Dimension Reduction.
Figure 4.
Figure 4.
Reparative peripheral nerve tissue shows RNA expression of anti-apoptotic factors across multiple cell types. (A) Anti-apoptosis factors were expressed broadly and robustly in many of the cell types of the reparative nerve for factors of interest: NFE2L2 (NRF2), BCL2, BCL2L1 (Bcl-xl), BCL6, and MCL1 of reparative peripheral nerve tissue. Violin plots show the relative expression across cell types (log2 average). Cell type key: SC: Schwann cell; mySC: myelinating Schwann cells; EC: endothelial cells; PC: proliferating cells; HC: heterogeneous cells; EMC: endoneurial mesenchymal cells; MC: mesenchymal cells; M: macrophages; PC/VMSC: pericytes/vascular smooth muscle cells; TC: T-cells. (B) Percentage of total cells expressing each anti-apoptotic factors. Note that the x-axis maximum is 40%. Data obtained from N = 2 participants.
Figure 5.
Figure 5.
Protein content of neuroprotective factors and anti-apoptotic factors. The mean protein concentration (and SD) in regenerating peripheral nerve tissue samples is summarized. BDNF (n = 15 participant samples), GDNF (n = 7), beta-NGF (n = 12), PDGFB (n = 15), VEGF (n = 15), NGFR (n = 14), CDNF (n = 15), PDGFA (n = 13), EPO (n = 13), NFE2L2 (n = 14), and BCL-6 (n = 15). BDNF: brain-derived neurotrophic factor; CDNF: cerebral dopamine neurotrophic factor; EPO: erythropoietin; GDNF: glial-cell derived neurotrophic factor; NGF: nerve growth factor; NGFR: nerve growth factor receptor; PDGFA: platelet-derived growth factor–A; PDGFB: platelet-derived growth factor–B; VEGF: vascular endothelial growth factor.
Figure 6.
Figure 6.
Proposed action of reparative peripheral nerve tissue transplant. Reparative peripheral nerve tissue deployed into the substantia nigra of participants with PD may act in multi-factorial ways with paracrine effects on the surrounding tissue. Anti-apoptotic factors may contribute to graft survival. Through this combination, a diversity of cell-types from regenerating peripheral nerve tissue could provide neuroprotective, pro-regenerative, and anti-inflammatory factors interacting with the degenerating cells in the CNS. Created with Biorender.com. CNS: central nervous system; PD: Parkinson’s disease.

References

    1. Olanow CW, Goetz CG, Kordower JH, Stoessl AJ, Sossi V, Brin MF, Shannon KM, Nauert GM, Perl DP, Godbold J, Freeman TB. A double-blind controlled trial of bilateral fetal nigral transplantation in Parkinson’s disease. Ann Neurol. 2003;54(3):403–14.
    1. Freed CR, Greene PE, Breeze RE, Tsai WY, DuMouchel W, Kao R, Dillon S, Winfield H, Culver S, Trojanowski JQ, Eidelberg D, et al.. Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. N Engl J Med. 2001;344(10):710–19.
    1. Wider C, Pollo C, Bloch J, Burkhard PR, Vingerhoets FJ. Long-term outcome of 50 consecutive Parkinson’s disease patients treated with subthalamic deep brain stimulation. Parkinsonism Relat Disord. 2008;14(2):114–19.
    1. Barker RA, TRANSEURO consortium. Designing stem-cell-based dopamine cell replacement trials for Parkinson’s disease. Nat Med. 2019;25(7):1045–53.
    1. Takahashi J. Preparing for first human trial of induced pluripotent stem cell-derived cells for Parkinson’s disease: an interview with Jun Takahashi. Regen Med. 2019;14(2):93–95.
    1. Araki R, Uda M, Hoki Y, Sunayama M, Nakamura M, Ando S, Sugiura M, Ideno H, Shimada A, Nifuji A, Abe M. Negligible immunogenicity of terminally differentiated cells derived from induced pluripotent or embryonic stem cells. Nature. 2013;494(7435):100–104.
    1. Zhao T, Zhang ZN, Rong Z, Xu Y. Immunogenicity of induced pluripotent stem cells. Nature. 2011;474(7350):212–15.
    1. van Horne CG, Quintero JE, Gurwell JA, Wagner RP, Slevin JT, Gerhardt GA. Implantation of autologous peripheral nerve grafts into the substantia Nigra of subjects with idiopathic Parkinson’s disease treated with bilateral STN DBS: a report of safety and feasibility. J Neurosurg. 2017;126(4):1140–47.
    1. van Horne CG, Quintero JE, Slevin JT, Anderson-Mooney A, Gurwell JA, Welleford AS, Lamm JR, Wagner RP, Gerhardt GA. Peripheral nerve grafts implanted into the substantia nigra in patients with Parkinson’s disease during deep brain stimulation surgery: 1-year follow-up study of safety, feasibility, and clinical outcome. J Neurosurg. 2018;129(6):1550–61.
    1. Weiss T, Taschner-Mandl S, Bileck A, Slany A, Kromp F, Rifatbegovic F, Frech C, Windhager R, Kitzinger H, Tzou CH, Ambros PF, et al.. Proteomics and transcriptomics of peripheral nerve tissue and cells unravel new aspects of the human Schwann cell repair phenotype. Glia. 2016;64(12):2133–53.
    1. Stierli S, Napoli I, White IJ, Cattin AL, Monteza Cabrejos A, Garcia Calavia N, Malong L, Ribeiro S, Nihouarn J, Williams R, Young KM, et al.. The regulation of the homeostasis and regeneration of peripheral nerve is distinct from the CNS and independent of a stem cell population. Development. 2018; 145(24):dev170316.
    1. Gordon T. Peripheral nerve regeneration and muscle reinnervation. Int J Mol Sci. 2020;21(22):8652.
    1. Welleford AS, Quintero JE, Seblani NE, Blalock E, Gunewardena S, Shapiro SM, Riordan SM, Huettl P, Guduru Z, Stanford JA, van Horne CG, et al.. RNA sequencing of human peripheral nerve in response to injury: distinctive analysis of the nerve repair pathways. Cell Transplant. 2020;29:963689720926157.
    1. Acheson A, Conover JC, Fandl JP, DeChiara TM, Russell M, Thadani A, Squinto SP, Yancopoulos GD, Lindsay RM. A BDNF autocrine loop in adult sensory neurons prevents cell death. Nature. 1995;374(6521):450–53.
    1. Chau MJ, Quintero JE, Blalock E, Samaan C, Gerhardt G, van Horne C. Transection injury differentially alters the proteome of the human sural nerve. Biorxiv. 2021:2021;20211123469670.
    1. Meyer M, Matsuoka I, Wetmore C, Olson L, Thoenen H. Enhanced synthesis of brain-derived neurotrophic factor in the lesioned peripheral nerve: different mechanisms are responsible for the regulation of BDNF and NGF mRNA. J Cell Biol. 1992;119(1):45–54.
    1. Hilton DA, Jacob J, Househam L, Tengah C. Complications following sural and peroneal nerve biopsies. J Neurol Neurosurg Psychiatry. 2007;78(11):1271–72.
    1. Campbell CA, Turza KC, Morgan RF. Postoperative outcomes and reliability of “sensation-sparing” sural nerve biopsy. Muscle Nerve. 2009;40(4):603–609.
    1. Gill SS, Patel NK, Hotton GR, O’Sullivan K, McCarter R, Bunnage M, Brooks DJ, Svendsen CN, Heywood P. Direct brain infusion of glial cell line-derived neurotrophic factor in Parkinson disease. Nat Med. 2003;9(5):589–95.
    1. Nutt JG, Burchiel KJ, Comella CL, Jankovic J, Lang AE, Laws ER, Jr, Lozano AM, Penn RD, Simpson RK, Jr, Stacy M, Wooten GF. Randomized, double-blind trial of glial cell line-derived neurotrophic factor (GDNF) in PD. Neurology. 2003;60(1):69–73.
    1. Collier TJ, Elsworth JD, Taylor JR, Sladek JR, Jr, Roth RH, Redmond DE., Jr. Peripheral nerve-dopamine neuron co-grafts in MPTP-treated monkeys: augmentation of tyrosine hydroxylase-positive fiber staining and dopamine content in host systems. Neuroscience. 1994;61(4):875–89.
    1. Collier TJ, Martin PN. Schwann cells as a source of neurotrophic activity for dopamine neurons. Exp Neurol. 1993;124(1):129–33.
    1. Bartus RT, Baumann TL, Siffert J, Herzog CD, Alterman R, Boulis N, Turner DA, Stacy M, Lang AE, Lozano AM, Olanow CW. Safety/feasibility of targeting the substantia nigra with AAV2-neurturin in Parkinson patients. Neurology. 2013;80(18):1698–1701.
    1. Whone A, Luz M, Boca M, Woolley M, Mooney L, Dharia S, Broadfoot J, Cronin D, Schroers C, Barua NU, Longpre L, et al.. Randomized trial of intermittent intraputamenal glial cell line-derived neurotrophic factor in Parkinson’s disease. Brain. 2019;142(3):512–25.
    1. Warren Olanow C, Bartus RT, Baumann TL, Factor S, Boulis N, Stacy M, Turner DA, Marks W, Larson P, Starr PA, Jankovic J, et al.. Gene delivery of neurturin to putamen and substantia nigra in Parkinson disease: a double-blind, randomized, controlled trial. Ann Neurol. 2015;78(2):248–57.
    1. Park KI. Transplantation of neural stem cells: cellular & gene therapy for hypoxic-ischemic brain injury. Yonsei Med J. 2000;41(6):825–35.
    1. Ishibashi S, Sakaguchi M, Kuroiwa T, Yamasaki M, Kanemura Y, Shizuko I, Shimazaki T, Onodera M, Okano H, Mizusawa H. Human neural stem/progenitor cells, expanded in long-term neurosphere culture, promote functional recovery after focal ischemia in Mongolian gerbils. J Neurosci Res. 2004;78(2):215–23.
    1. Kelly S, Bliss TM, Shah AK, Sun GH, Ma M, Foo WC, Masel J, Yenari MA, Weissman IL, Uchida N, Palmer T, et al.. Transplanted human fetal neural stem cells survive, migrate, and differentiate in ischemic rat cerebral cortex. Proc Natl Acad Sci U S A. 2004;101(32):11839–44.
    1. Carrington EM, Zhan Y, Brady JL, Zhang JG, Sutherland RM, Anstee NS, Schenk RL, Vikstrom IB, Delconte RB, Segal D, Huntington ND, et al.. Anti-apoptotic proteins BCL-2, MCL-1 and A1 summate collectively to maintain survival of immune cell populations both in vitro and in vivo. Cell Death Differ. 2017;24(5):878–88.
    1. Carriel V, Garzón I, Alaminos M, Campos A. Evaluation of myelin sheath and collagen reorganization pattern in a model of peripheral nerve regeneration using an integrated histochemical approach. Histochem Cell Biol. 2011;136(6):709–17.
    1. Raimondo S, Fornaro M, Di Scipio F, Ronchi G, Giacobini-Robecchi MG, Geuna S. Chapter 5: methods and protocols in peripheral nerve regeneration experimental research: part II-morphological techniques. Int Rev Neurobiol. 2009;87:81–103.
    1. Carr MJ, Toma JS, Johnston APW, Steadman PE, Yuzwa SA, Mahmud N, Frankland PW, Kaplan DR, Miller FD. Mesenchymal precursor cells in adult nerves contribute to mammalian tissue repair and regeneration. Cell Stem Cell. 2019;24(2):240.e9–56.
    1. Chen B, Banton MC, Singh L, Parkinson DB, Dun XP. Single cell transcriptome data analysis defines the heterogeneity of peripheral nerve cells in homeostasis and regeneration. Front Cell Neurosci. 2021;15:624826.
    1. Li M, Min Q, Banton MC, Dun X. Single-cell regulatory network inference and clustering identifies cell-type specific expression pattern of transcription factors in mouse sciatic nerve. Front Cell Neurosci. 2021;15:676515.
    1. Richard L, Vedrenne N, Vallat JM, Funalot B. Characterization of endoneurial fibroblast-like cells from human and rat peripheral nerves. J Histochem Cytochem. 2014;62(6):424–35.
    1. Clements MP, Byrne E, Camarillo Guerrero LF, Cattin AL, Zakka L, Ashraf A, Burden JJ, Khadayate S, Lloyd AC, Marguerat S, Parrinello S. The wound microenvironment reprograms Schwann cells to invasive mesenchymal-like cells to drive peripheral nerve regeneration. Neuron. 2017;96(1):98.e7–14.
    1. Peng K, Sant D, Andersen N, Silvera R, Camarena V, Pinero G, Graham R, Khan A, Xu XM, Wang G, Monje PV. Magnetic separation of peripheral nerve-resident cells underscores key molecular features of human Schwann cells and fibroblasts: an immunochemical and transcriptomics approach. Sci Rep. 2020;10(1):18433.
    1. Wei Z, Shu S, Zhang M, Xie S, Tang S, Nie K, Li H. A subpopulation of Schwann cell-like cells with nerve regeneration signatures is identified through single-cell RNA sequencing. Front Physiol. 2021;12:637924.
    1. Gerber D, Pereira JA, Gerber J, Tan G, Dimitrieva S, Yanguez E, Suter U. Transcriptional profiling of mouse peripheral nerves to the single-cell level to build a sciatic nerve ATlas (SNAT). Elife. 2021;10:e58591.
    1. Toma JS, Karamboulas K, Carr MJ, Kolaj A, Yuzwa SA, Mahmud N, Storer MA, Kaplan DR, Miller FD. Peripheral nerve single-cell analysis identifies mesenchymal ligands that promote axonal growth. Eneuro. 2020;7(3):2020.
    1. Wolbert J, Li X, Heming M, Mausberg AK, Akkermann D, Frydrychowicz C, Fledrich R, Groeneweg L, Schulz C, Stettner M, Gonzalez NA, et al.. Redefining the heterogeneity of peripheral nerve cells in health and autoimmunity. Proc Natl Acad Sci U S A. 2020;117(17):9466–76.
    1. Miller BA, Turan N, Chau M, Pradilla G. Inflammation, vasospasm, and brain injury after subarachnoid hemorrhage. Biomed Res Int. 2014;2014:384342.
    1. Pellegrino RG, Politis MJ, Ritchie JM, Spencer PS. Events in degenerating cat peripheral nerve: induction of Schwann cell S phase and its relation to nerve fibre degeneration. J Neurocytol. 1986;15(1):17–28.
    1. Arthur-Farraj PJ, Latouche M, Wilton DK, Quintes S, Chabrol E, Banerjee A, Woodhoo A, Jenkins B, Rahman M, Turmaine M, Wicher GK, et al.. c-Jun reprograms Schwann cells of injured nerves to generate a repair cell essential for regeneration. Neuron. 2012;75(4):633–47.
    1. Fontana X, Hristova M, Da Costa C, Patodia S, Thei L, Makwana M, Spencer-Dene B, Latouche M, Mirsky R, Jessen KR, Klein R, et al.. c-Jun in Schwann cells promotes axonal regeneration and motoneuron survival via paracrine signaling. J Cell Biol. 2012;198(1):127–41.
    1. Jessen KR, Mirsky R. The repair Schwann cell and its function in regenerating nerves. J Physiol. 2016;594(13):3521–31.
    1. La Fleur M, Underwood JL, Rappolee DA, Werb Z. Basement membrane and repair of injury to peripheral nerve: defining a potential role for macrophages, matrix metalloproteinases, and tissue inhibitor of metalloproteinases-1. J Exp Med. 1996;184(6):2311–26.
    1. Mueller M, Leonhard C, Wacker K, Ringelstein EB, Okabe M, Hickey WF, Kiefer R. Macrophage response to peripheral nerve injury: the quantitative contribution of resident and hematogenous macrophages. Lab Invest. 2003;83(2):175–85.
    1. Stratton JA, Holmes A, Rosin NL, Sinha S, Vohra M, Burma NE, Trang T, Midha R, Biernaskie J. Macrophages regulate Schwann cell maturation after nerve injury. Cell Rep. 2018; 24(10):2561.e6–72.e6.
    1. Wilcox MB, Laranjeira SG, Eriksson TM, Jessen KR, Mirsky R, Quick TJ, Phillips JB. Characterising cellular and molecular features of human peripheral nerve degeneration. Acta Neuropathol Commun. 2020;8(1):51.
    1. Anderson KD, Guest JD, Dietrich WD, Bartlett Bunge M, Curiel R, Dididze M, Green BA, Khan A, Pearse DD, Saraf-Lavi E, Widerström-Noga E. Safety of autologous human Schwann cell transplantation in subacute thoracic spinal cord injury. J Neurotrauma. 2017;34(21):2950–63.
    1. Xia Y, Jiang C, Cao Z, Shi K, Wang Y. Co-transplantation of macaque autologous Schwann cells and human embryonic nerve stem cells in treatment of macaque Parkinson’s disease. Asian Pac J Trop Med. 2012;5(1):7–14.
    1. Quintero JE, Slevin JT, Gurwell JA, McLouth CJ, Khouli RE, Chau MJ, Guduru Z, Gerhardt GA, van Horne CG. Direct delivery of an investigational cell therapy in patients with Parkinson’s disease: an interim analysis of feasibility and safety of an open-label study using DBS-Plus clinical trial design. BMJ Neurology Open 2022;4:e000301.
    1. Jessen KR, Mirsky R. The success and failure of the Schwann cell response to nerve injury. Front Cell Neurosci. 2019;13:33.
    1. Jessen KR, Mirsky R, Lloyd AC. Schwann cells: development and role in nerve repair. Cold Spring Harb Perspect Biol. 2015;7(7):a020487.
    1. Jessen KR, Arthur-Farraj P. Repair Schwann cell update: adaptive reprogramming, EMT, and stemness in regenerating nerves. Glia. 2019;67(3):421–37.
    1. Allodi I, Udina E, Navarro X. Specificity of peripheral nerve regeneration: interactions at the axon level. Prog Neurobiol. 2012;98(1):16–37.
    1. Blesch A, Lu P, Tsukada S, Alto LT, Roet K, Coppola G, Geschwind D, Tuszynski MH. Conditioning lesions before or after spinal cord injury recruit broad genetic mechanisms that sustain axonal regeneration: superiority to camp-mediated effects. Exp Neurol. 2012;235(1):162–73.
    1. Roberson MD, Toews AD, Bouldin TW, Weaver J, Goines ND, Morell P. NGFR-mRNA expression in sciatic nerve: a sensitive indicator of early stages of axonopathy. Brain Res Mol Brain Res. 1995;28(2):231–38.
    1. Rosenberg AF, Wolman MA, Franzini-Armstrong C, Granato M. In vivo nerve-macrophage interactions following peripheral nerve injury. J Neurosci. 2012;32(11):3898–3909.
    1. Gensel JC, Zhang B. Macrophage activation and its role in repair and pathology after spinal cord injury. Brain Res. 2015;1619:1–11.
    1. Ferrante CJ, Leibovich SJ. Regulation of macrophage polarization and wound healing. Adv Wound Care (New Rochelle). 2012;1(1):10–16.
    1. Chen P, Piao X, Bonaldo P. Role of macrophages in Wallerian degeneration and axonal regeneration after peripheral nerve injury. Acta Neuropathol. 2015;130(5):605–18.
    1. Tsuyuzaki K, Sato H, Sato K, Nikaido I. Benchmarking principal component analysis for large-scale single-cell RNA-sequencing. Genome Biol. 2020;21(1):9.
    1. Li W, Englund E, Widner H, Mattsson B, van Westen D, Lätt J, Rehncrona S, Brundin P, Björklund A, Lindvall O, Li J-Y. Extensive graft-derived dopaminergic innervation is maintained 24 years after transplantation in the degenerating parkinsonian brain. Proceedings of the National Academy of Sciences. 2016;113(23):6544.
    1. Lindvall O. Clinical translation of stem cell transplantation in Parkinson’s disease. J Intern Med. 2016;279(1):30–40.
    1. Olanow CW, Freeman T, Kordower J. Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. N Engl J Med 2001;345(2):146.
    1. Mendez I, Vinuela A, Astradsson A, Mukhida K, Hallett P, Robertson H, Tierney T, Holness R, Dagher A, Trojanowski JQ, Isacson O. Dopamine neurons implanted into people with Parkinson’s disease survive without pathology for 14 years. Nat Med. 2008;14(5):507–509.
    1. Grambalova Z, Kaiserova M, Vastik M, Mensikova K, Otruba P, Zapletalova J, Dufek J, Kanovsky P. Peripheral neuropathy in Parkinson’s disease. Neuro Endocrinol Lett. 2015;36(4):363–67.
    1. Zis P, Grunewald RA, Chaudhuri RK, Hadjivassiliou M. Peripheral neuropathy in idiopathic Parkinson’s disease: a systematic review. J Neurol Sci. 2017;378:204–209.
    1. Chau MJ, Deveau TC, Gu X, Kim YS, Xu Y, Yu SP, Wei L. Delayed and repeated intranasal delivery of bone marrow stromal cells increases regeneration and functional recovery after ischemic stroke in mice. BMC Neurosci. 2018; 19(1):20.
    1. Chau MJ, Deveau TC, Song M, Gu X, Chen D, Wei L. iPSC Transplantation increases regeneration and functional recovery after ischemic stroke in neonatal rats. Stem Cells. 2014;32(12):3075–87.
    1. Chau M, Deveau TC, Song M, Wei ZZ, Gu X, Yu SP, Wei L. Transplantation of iPS cell-derived neural progenitors overexpressing SDF-1alpha increases regeneration and functional recovery after ischemic stroke. Oncotarget. 2017;8(57):97537–53.

Source: PubMed

3
Abonnieren