1
|
Kumar S, Kabat M, Basak S, Babiarz J, Berthiaume F, Grumet M. Anti-Inflammatory Effects of Encapsulated Human Mesenchymal Stromal/Stem Cells and a Method to Scale-Up Cell Encapsulation. Biomolecules 2022; 12:biom12121803. [PMID: 36551231 PMCID: PMC9775968 DOI: 10.3390/biom12121803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSC) promote recovery in a wide range of animal models of injury and disease. They can act in vivo by differentiating and integrating into tissues, secreting factors that promote cell growth and control inflammation, and interacting directly with host effector cells. We focus here on MSC secreted factors by encapsulating the cells in alginate microspheres, which restrict cells from migrating out while allowing diffusion of factors including cytokines across the capsules. One week after intrathecal lumbar injection of human bone marrow MSC encapsulated in alginate (eMSC), rat IL-10 expression was upregulated in distant rat spinal cord injury sites. Detection of human IL-10 protein in rostrally derived cerebrospinal fluid (CSF) indicated distribution of this human MSC-secreted cytokine throughout rat spinal cord CSF. Intraperitoneal (IP) injection of eMSC in a rat model for endotoxemia reduced serum levels of inflammatory cytokines within 5 h. Detection of human IL-6 in sera after injection of human eMSC indicates rapid systemic distribution of this human MSC-secreted cytokine. Despite proof of concept for eMSC in various disorders using animal models, translation of encapsulation technology has not been feasible primarily because methods for scale-up are not available. To scale-up production of eMSC, we developed a rapid, semi-continuous, capsule collection system coupled to an electrosprayer. This system can produce doses of encapsulated cells sufficient for use in clinical translation.
Collapse
Affiliation(s)
- Suneel Kumar
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- W. M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Maciej Kabat
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- W. M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Sayantani Basak
- W. M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Joanne Babiarz
- W. M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Francois Berthiaume
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Martin Grumet
- W. M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Correspondence: ; Tel.: +1-917-597-2597; Fax: +1-732-445-2063
| |
Collapse
|
2
|
Hall A, Fortino T, Spruance V, Niceforo A, Harrop JS, Phelps PE, Priest CA, Zholudeva LV, Lane MA. Cell transplantation to repair the injured spinal cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:79-158. [PMID: 36424097 PMCID: PMC10008620 DOI: 10.1016/bs.irn.2022.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Adam Hall
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Tara Fortino
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Victoria Spruance
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Division of Kidney, Urologic, & Hematologic Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alessia Niceforo
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - James S Harrop
- Department of Neurological and Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patricia E Phelps
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, United States
| | | | - Lyandysha V Zholudeva
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Gladstone Institutes, San Francisco, CA, United States
| | - Michael A Lane
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
3
|
Abstract
Spinal cord injury represents a devastating central nervous system injury that could impair the mobility and sensory function of afflicted patients. The hallmarks of spinal cord injury include neuroinflammation, axonal degeneration, neuronal loss, and reactive gliosis. Furthermore, the formation of a glial scar at the injury site elicits an inhibitory environment for potential neuroregeneration. Besides axonal regeneration, a significant challenge in treating spinal cord injury is to replenish the neurons lost during the pathological process. However, despite decades of research efforts, current strategies including stem cell transplantation have not resulted in a successful clinical therapy. Furthermore, stem cell transplantation faces serious hurdles such as immunorejection of the transplanted cells and ethical issues. In vivo neuronal reprogramming is a recently developed technology and leading a major breakthrough in regenerative medicine. This innovative technology converts endogenous glial cells into functional neurons for injury repair in the central nervous system. The feasibility of in vivo neuronal reprogramming has been demonstrated successfully in models of different neurological disorders including spinal cord injury by numerous laboratories. Several reprogramming factors, mainly the pro-neural transcription factors, have been utilized to reprogram endogenous glial cells into functional neurons with distinct phenotypes. So far, the literature on in vivo neuronal reprogramming in the model of spinal cord injury is still small. In this review, we summarize a limited number of such reports and discuss several questions that we think are important for applying in vivo neuronal reprogramming in the research field of spinal cord injury as well as other central nervous system disorders.
Collapse
Affiliation(s)
- Xuanyu Chen
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Hedong Li
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| |
Collapse
|
4
|
Llorente IL, Hatanaka EA, Meadow ME, Xie Y, Lowry WE, Carmichael ST. Reliable generation of glial enriched progenitors from human fibroblast-derived iPSCs. Stem Cell Res 2021; 55:102458. [PMID: 34274773 PMCID: PMC8444576 DOI: 10.1016/j.scr.2021.102458] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 05/06/2021] [Accepted: 07/05/2021] [Indexed: 12/15/2022] Open
Abstract
White matter stroke (WMS) occurs as small infarcts in deep penetrating blood vessels in the brain and affects the regions of the brain that carry connections, termed the subcortical white matter. WMS progresses over years and has devastating clinical consequences. Unlike large grey matter strokes, WMS disrupts the axonal architecture of the brain and depletes astrocytes, oligodendrocyte lineage cells, axons and myelinating cells, resulting in abnormalities of gait and executive function. An astrocytic cell-based therapy is positioned as a strong therapeutic candidate after WMS. In this study we report, the reliable generation of a novel stem cell-based therapeutic product, glial enriched progenitors (GEPs) derived from human induced pluripotent stem cells (hiPSCs). By transient treatment of hiPSC derived neural progenitors (hiPSC-NPCs) with the small molecule deferoxamine, a prolyl hydroxylase inhibitor, for three days hiPSC-NPCs become permanently biased towards an astrocytic fate, producing hiPSC-GEPs. In preparation for clinical application, we have developed qualification assays to ensure identity, safety, purity, and viability of the cells prior to manufacture. Using tailored q-RT-PCR-based assays, we have demonstrated the lack of pluripotency in our final therapeutic candidate cells (hiPSC-GEPs) and we have identified the unique genetic profile of hiPSC-GEPs that is clearly distinct from the parent lines, hiPSCs and iPSC-NPCs. After completion of the viability assay, we have stablished the therapeutic window of use for hiPSC-GEPs in future clinical applications (7 h). Lastly, we were able to reliably and consistently produce a safe therapeutic final product negative for contamination by any human or murine viral pathogens, selected bacteria, common laboratory mycoplasmas, growth of any aerobes, anaerobes, yeast, or fungi and 100 times less endotoxin levels than the maximum acceptable value. This study demonstrates the reliable and safe generation of patient derived hiPSC-GEPs that are clinically ready as a cell-based therapeutic approach for WMS.
Collapse
Affiliation(s)
- Irene L Llorente
- Department of Neurology, David Geffen School of Medicine at UCLA, USA
| | - Emily A Hatanaka
- Department of Molecular, Cell and Developmental Biology, UCLA, USA
| | - Michael E Meadow
- Department of Molecular, Cell and Developmental Biology, UCLA, USA
| | - Yuan Xie
- Department of Biochemistry and Molecular Biology, University of Chicago, USA
| | - William E Lowry
- Department of Molecular, Cell and Developmental Biology, UCLA, USA
| | | |
Collapse
|
5
|
Li X, Gao Y, Tian F, Du R, Yuan Y, Li P, Liu F, Wang C. miR-31 promotes neural stem cell proliferation and restores motor function after spinal cord injury. Exp Biol Med (Maywood) 2021; 246:1274-1286. [PMID: 33715531 PMCID: PMC8371310 DOI: 10.1177/1535370221997071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 02/01/2021] [Indexed: 01/17/2023] Open
Abstract
This study aims to examine whether miR-31 promotes endogenous NSC proliferation and be used for spinal cord injury management. In the present study, the morpholino knockdown of miR-31 induced abnormal neuronal apoptosis in zebrafish, resulting in impaired development of the tail. miR-31 agomir transfection in NSCs increased Nestin expression and decreased ChAT and GFAP expression levels. miR-31 induced the proliferation of mouse NSCs by upregulating the Notch signaling pathway, and more NSCs entered G1; Notch was inhibited by miR-31 inactivation. Injection of a miR-31 agomir into mouse models of spinal cord injury could effectively restore motor functions after spinal cord injury, which was achieved by promoting the proliferation of endogenous NSCs. After the injection of a miR-31 agomir in spinal cord injury mice, the expression of Nestin and GFAP increased, while GFAP expression decreased. In conclusion, the zebrafish experiments prove that a lack of miR-31 will block nervous system development. In spinal cord injury mouse models, miR-31 overexpression might promote spinal cord injury repair.
Collapse
Affiliation(s)
- Xiao Li
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Yuantao Gao
- Queen Mary School, Nanchang University, Nanchang 330000, China
| | - Feng Tian
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Ruochen Du
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Yitong Yuan
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Pengfei Li
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Fang Liu
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| | - Chunfang Wang
- Laboratory Animal Research Center of Shanxi Medical University, Shanxi Key Laboratory of Animal and Animal Model of Human Diseases, Taiyuan 030001, China
| |
Collapse
|
6
|
Beyer F, Samper Agrelo I, Küry P. Do Neural Stem Cells Have a Choice? Heterogenic Outcome of Cell Fate Acquisition in Different Injury Models. Int J Mol Sci 2019; 20:ijms20020455. [PMID: 30669690 PMCID: PMC6359747 DOI: 10.3390/ijms20020455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/14/2019] [Accepted: 01/18/2019] [Indexed: 12/19/2022] Open
Abstract
The adult mammalian central nervous system (CNS) is generally considered as repair restricted organ with limited capacities to regenerate lost cells and to successfully integrate them into damaged nerve tracts. Despite the presence of endogenous immature cell types that can be activated upon injury or in disease cell replacement generally remains insufficient, undirected, or lost cell types are not properly generated. This limitation also accounts for the myelin repair capacity that still constitutes the default regenerative activity at least in inflammatory demyelinating conditions. Ever since the discovery of endogenous neural stem cells (NSCs) residing within specific niches of the adult brain, as well as the description of procedures to either isolate and propagate or artificially induce NSCs from various origins ex vivo, the field has been rejuvenated. Various sources of NSCs have been investigated and applied in current neuropathological paradigms aiming at the replacement of lost cells and the restoration of functionality based on successful integration. Whereas directing and supporting stem cells residing in brain niches constitutes one possible approach many investigations addressed their potential upon transplantation. Given the heterogeneity of these studies related to the nature of grafted cells, the local CNS environment, and applied implantation procedures we here set out to review and compare their applied protocols in order to evaluate rate-limiting parameters. Based on our compilation, we conclude that in healthy CNS tissue region specific cues dominate cell fate decisions. However, although increasing evidence points to the capacity of transplanted NSCs to reflect the regenerative need of an injury environment, a still heterogenic picture emerges when analyzing transplantation outcomes in injury or disease models. These are likely due to methodological differences despite preserved injury environments. Based on this meta-analysis, we suggest future NSC transplantation experiments to be conducted in a more comparable way to previous studies and that subsequent analyses must emphasize regional heterogeneity such as accounting for differences in gray versus white matter.
Collapse
Affiliation(s)
- Felix Beyer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, D-40225 Düsseldorf, Germany.
| | - Iria Samper Agrelo
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, D-40225 Düsseldorf, Germany.
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, D-40225 Düsseldorf, Germany.
| |
Collapse
|
7
|
Skop NB, Singh S, Antikainen H, Saqcena C, Calderon F, Rothbard DE, Cho CH, Gandhi CD, Levison SW, Dobrowolski R. Subacute Transplantation of Native and Genetically Engineered Neural Progenitors Seeded on Microsphere Scaffolds Promote Repair and Functional Recovery After Traumatic Brain Injury. ASN Neuro 2019; 11:1759091419830186. [PMID: 30818968 PMCID: PMC6399762 DOI: 10.1177/1759091419830186] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/11/2018] [Accepted: 12/18/2018] [Indexed: 01/09/2023] Open
Abstract
There is intense interest and effort toward regenerating the brain after severe injury. Stem cell transplantation after insult to the central nervous system has been regarded as the most promising approach for repair; however, engrafting cells alone might not be sufficient for effective regeneration. In this study, we have compared neural progenitors (NPs) from the fetal ventricular zone (VZ), the postnatal subventricular zone, and an immortalized radial glia (RG) cell line engineered to conditionally secrete the trophic factor insulin-like growth factor 1 (IGF-1). Upon differentiation in vitro, the VZ cells were able to generate a greater number of neurons than subventricular zone cells. Furthermore, differentiated VZ cells generated pyramidal neurons . In vitro, doxycycline-driven secretion of IGF-1 strongly promoted neuronal differentiation of cells with hippocampal, interneuron and cortical specificity. Accordingly, VZ and engineered RG-IGF-1-hemagglutinin (HA) cells were selected for subsequent in vivo experiments. To increase cell survival, we delivered the NPs attached to a multifunctional chitosan-based scaffold. The microspheres containing adherent NPs were injected subacutely into the lesion cavity of adult rat brains that had sustained controlled cortical impact injury. At 2 weeks posttransplantation, the exogenously introduced cells showed a reduction in stem cell or progenitor markers and acquired mature neuronal and glial markers. In beam walking tests assessing sensorimotor recovery, transplanted RG cells secreting IGF-1 contributed significantly to functional improvement while native VZ or RG cells did not promote significant recovery. Altogether, these results support the therapeutic potential of chitosan-based multifunctional microsphere scaffolds seeded with genetically modified NPs expressing IGF-1 to promote repair and functional recovery after traumatic brain injuries.
Collapse
Affiliation(s)
- Nolan B. Skop
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, NJ, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Sweta Singh
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
- Stem Cell and Gene Therapy Research Group, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Henri Antikainen
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Chaitali Saqcena
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
| | - Frances Calderon
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, NJ, USA
| | - Deborah E. Rothbard
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, NJ, USA
| | - Cheul H. Cho
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, USA
| | - Chirag D. Gandhi
- Department of Neurosurgery, Westchester Medical Center at NY Medical College, Valhalla, NY, USA
| | - Steven W. Levison
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, NJ, USA
| | - Radek Dobrowolski
- Department of Biological Sciences, Rutgers University, Newark, NJ, USA
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, TX, USA
| |
Collapse
|
8
|
Mashanov V, Zueva O. Radial Glia in Echinoderms. Dev Neurobiol 2018; 79:396-405. [PMID: 30548565 DOI: 10.1002/dneu.22659] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 11/13/2018] [Accepted: 11/28/2018] [Indexed: 12/12/2022]
Abstract
Radial glial cells are crucial in vertebrate neural development and regeneration. It has been recently proposed that this neurogenic cell type might be older than the chordate lineage itself and might have been present in the last common deuterostome ancestor. Here, we summarize the results of recent studies on radial glia in echinoderms, a highly regenerative phylum of marine invertebrates with shared ancestry to chordates. We discuss the involvement of these cells in both homeostatic neurogenesis and post-traumatic neural regeneration, compare the features of radial glia in echinoderms and chordates to each other, and review the molecular mechanisms that control differentiation and plasticity of the echinoderm radial glia. Overall, studies on echinoderm radial glia provide a unique opportunity to understand the fundamental biology of this cell type from evolutionary and comparative perspectives.
Collapse
Affiliation(s)
- Vladimir Mashanov
- Department of Biology, University of North Florida, Jacksonville, Florida
| | - Olga Zueva
- Department of Biology, University of North Florida, Jacksonville, Florida.,Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, Pennsylvania
| |
Collapse
|
9
|
Wollenberg AL, O'Shea TM, Kim JH, Czechanski A, Reinholdt LG, Sofroniew MV, Deming TJ. Injectable polypeptide hydrogels via methionine modification for neural stem cell delivery. Biomaterials 2018; 178:527-545. [PMID: 29657091 PMCID: PMC6054810 DOI: 10.1016/j.biomaterials.2018.03.057] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/11/2018] [Accepted: 03/31/2018] [Indexed: 12/21/2022]
Abstract
Injectable hydrogels with tunable physiochemical and biological properties are potential tools for improving neural stem/progenitor cell (NSPC) transplantation to treat central nervous system (CNS) injury and disease. Here, we developed injectable diblock copolypeptide hydrogels (DCH) for NSPC transplantation that contain hydrophilic segments of modified l-methionine (Met). Multiple Met-based DCH were fabricated by post-polymerization modification of Met to various functional derivatives, and incorporation of different amino acid comonomers into hydrophilic segments. Met-based DCH assembled into self-healing hydrogels with concentration and composition dependent mechanical properties. Mechanical properties of non-ionic Met-sulfoxide formulations (DCHMO) were stable across diverse aqueous media while cationic formulations showed salt ion dependent stiffness reduction. Murine NSPC survival in DCHMO was equivalent to that of standard culture conditions, and sulfoxide functionality imparted cell non-fouling character. Within serum rich environments in vitro, DCHMO was superior at preserving NSPC stemness and multipotency compared to cell adhesive materials. NSPC in DCHMO injected into uninjured forebrain remained local and, after 4 weeks, exhibited an immature astroglial phenotype that integrated with host neural tissue and acted as cellular substrates that supported growth of host-derived axons. These findings demonstrate that Met-based DCH are suitable vehicles for further study of NSPC transplantation in CNS injury and disease models.
Collapse
Affiliation(s)
- A L Wollenberg
- Departments of Bioengineering, Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1600, USA
| | - T M O'Shea
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1763, USA
| | - J H Kim
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1763, USA
| | - A Czechanski
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | | | - M V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1763, USA
| | - T J Deming
- Departments of Bioengineering, Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095-1600, USA.
| |
Collapse
|
10
|
Spinal Cord Stem Cells In Their Microenvironment: The Ependyma as a Stem Cell Niche. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1041:55-79. [PMID: 29204829 DOI: 10.1007/978-3-319-69194-7_5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The ependyma of the spinal cord is currently proposed as a latent neural stem cell niche. This chapter discusses recent knowledge on the developmental origin and nature of the heterogeneous population of cells that compose this stem cell microenviroment, their diverse physiological properties and regulation. The chapter also reviews relevant data on the ependymal cells as a source of plasticity for spinal cord repair.
Collapse
|
11
|
Venkatachalam S, Neelamegan S, Okuda T, Marcus A, Woodbury D, Grumet M. Potential risk of clonally expanded amnion mesenchymal stem cell transplants in contused spinal cords. Restor Neurol Neurosci 2018; 36:387-396. [PMID: 29614703 DOI: 10.3233/rnn-170786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Mesenchymal stem/stromal cells (MSC) promote recovery after spinal cord injury (SCI) using adult bone marrow MSC (BM-MSC). Newborn tissues are a convenient source of MSC that does not involve an invasive procedure for cell collection. In this study the authors tested the effects of rat amnion MSC clone (rAM-MSC) in SCI. METHODS We tested intra-parenchymal injection of a GFP+ rat rAM-MSC clone derived from E18.5 rats in rat SCI and measured behavioral recovery (BBB scores), histology and X-ray opacity. Expression of aggrecan was measured in culture after treatment with TGFß. RESULTS Injection of rAM-MSC after SCI did not improve BBB scores compared to control vehicle injections; rather they reduced scores significantly over 6 weeks. Spinal cords injected with rAM-MSC were hard in regions surrounding the SCI site, which was confirmed by X-ray opacity. Whole mount imaging of these cords showed minimal tissue loss in the SCI site that occurred in SCI controls, and persistence of GFP+ rAM-MSC. Mason's Trichrome staining of tissue sections showed more intense staining for extracellular matrix (ECM) surrounding and extending beyond the SCI site with injections of rAM-MSC but not in controls. In response to TGF-ß treatment in culture, chondrogenic aggrecan was expressed at higher levels in rAM-MSC than in rBM-MSC, suggesting that the upregulation of TGF-ß in SCI sites may promote chondrogenic differentiation. CONCLUSION Acute injection after SCI of a clonally expanded rAM-MSC resulted in aberrant differentiation towards a chondrocytic phenotype that disrupts the spinal cord and inhibits behavioral recovery after SCI. It will be critical to ensure that injection of extensively expanded neonatal cells do not differentiate aberrantly in traumatic CNS tissue and disrupt recovery.
Collapse
Affiliation(s)
- Sankar Venkatachalam
- Department of Cell Biology and Neuroscience, W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA.,Department of Anatomy, University of Madras, Taramani Campus, Chennai, Tamilnadu, India
| | - Sridharan Neelamegan
- Department of Anatomy, University of Madras, Taramani Campus, Chennai, Tamilnadu, India
| | - Tetsuhito Okuda
- Department of Cell Biology and Neuroscience, W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Akiva Marcus
- The Ira B. Black Center for Stem Cell Research and The Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Dale Woodbury
- The Ira B. Black Center for Stem Cell Research and The Department of Neuroscience and Cell Biology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Martin Grumet
- Department of Cell Biology and Neuroscience, W. M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
12
|
Moore L, Skop NB, Rothbard DE, Corrubia LR, Levison SW. Tethered growth factors on biocompatible scaffolds improve stemness of cultured rat and human neural stem cells and growth of oligodendrocyte progenitors. Methods 2018; 133:54-64. [DOI: 10.1016/j.ymeth.2017.08.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/17/2017] [Accepted: 08/24/2017] [Indexed: 10/18/2022] Open
|
13
|
Marichal N, Reali C, Rehermann MI, Trujillo-Cenóz O, Russo RE. Progenitors in the Ependyma of the Spinal Cord: A Potential Resource for Self-Repair After Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1015:241-264. [DOI: 10.1007/978-3-319-62817-2_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
14
|
Wang H, Mei X, Cao Y, Liu C, Zhao Z, Guo Z, Bi Y, Shen Z, Yuan Y, Guo Y, Song C, Bai L, Wang Y, Yu D. HMGB1/Advanced Glycation End Products (RAGE) does not aggravate inflammation but promote endogenous neural stem cells differentiation in spinal cord injury. Sci Rep 2017; 7:10332. [PMID: 28871209 PMCID: PMC5583351 DOI: 10.1038/s41598-017-10611-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 08/10/2017] [Indexed: 12/31/2022] Open
Abstract
Receptor for advanced glycation end products (RAGE) signaling is involved in a series of cell functions after spinal cord injury (SCI). Our study aimed to elucidate the effects of RAGE signaling on the neuronal recovery after SCI. In vivo, rats were subjected to SCI with or without anti-RAGE antibodies micro-injected into the lesion epicenter. We detected Nestin/RAGE, SOX-2/RAGE and Nestin/MAP-2 after SCI by Western blot or immunofluorescence (IF). We found that neural stem cells (NSCs) co-expressed with RAGE were significantly activated after SCI, while stem cell markers Nestin and SOX-2 were reduced by RAGE blockade. We found that RAGE inhibition reduced nestin-positive NSCs expressing MAP-2, a mature neuron marker. RAGE blockade does not improve neurobehavior Basso, Beattie and Bresnahan (BBB) scores; however, it damaged survival of ventral neurons via Nissl staining. Through in vitro study, we found that recombinant HMGB1 administration does not lead to increased cytokines of TNF-α and IL-1β, while anti-RAGE treatment reduced cytokines of TNF-α and IL-1β induced by LPS via ELISA. Meanwhile, HMGB1 increased MAP-2 expression, which was blocked after anti-RAGE treatment. Hence, HMGB1/RAGE does not exacerbate neuronal inflammation but plays a role in promoting NSCs differentiating into mature neurons in the pathological process of SCI.
Collapse
Affiliation(s)
- Hongyu Wang
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Xifan Mei
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China.
| | - Yang Cao
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Chang Liu
- Department of Endocrinology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Ziming Zhao
- Department of Stomatology, Second Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Zhanpeng Guo
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Yunlong Bi
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Zhaoliang Shen
- Department of Orthopedics, Second Hospital of Jinzhou, Jinzhou City, PR China
| | - Yajiang Yuan
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Yue Guo
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Cangwei Song
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Liangjie Bai
- Department of Orthopedics, China Medical University, Shenyang City, PR China
| | - Yansong Wang
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| | - Deshui Yu
- Department of Orthopedic, First Affiliated Hospital of Jinzhou Medical University, Jinzhou City, PR China
| |
Collapse
|
15
|
Neural Stem Cell Transplantation Promotes Functional Recovery from Traumatic Brain Injury via Brain Derived Neurotrophic Factor-Mediated Neuroplasticity. Mol Neurobiol 2017; 55:2696-2711. [PMID: 28421542 DOI: 10.1007/s12035-017-0551-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 04/07/2017] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) induces cognitive impairments, motor and behavioral deficits. Previous evidences have suggested that neural stem cell (NSC) transplantation could facilitate functional recovery from brain insults, but their underlying mechanisms remains to be elucidated. Here, we established TBI model by an electromagnetic-controlled cortical impact device in the rats. Then, 5 μl NSCs (5.0 × 105/μl), derived from green fluorescent protein (GFP) transgenic mouse, was transplanted into the traumatic brain regions of rats at 24 h after injury. After differentiation of the NSCs was determined using immunohistochemistry, neurological severity scores (NSS) and rotarod test were conducted to detect the neurological behavior. Western blot and RT-PCR as well as ELASA were used to evaluate the expression of synaptophysin and brain-derived neurotrophic factor (BDNF). In order to elucidate the role of BDNF on the neural recovery after NSC transplantation, BDNF knockdown in NSC was performed and transplanted into the rats with TBI, and potential mechanism for BDNF knockdown in the NSC was analyzed using microassay analysis. Meanwhile, BDNF antibody blockade was conducted to further confirm the effect of BDNF on neural activity. As a result, an increasing neurological function improvement was seen in NSC transplanted rats, which was associated with the upregulation of synaptophysin and BDNF expression. Moreover, transplantation of BDNF knockdown NSCs and BDNF antibody block reduced not only the level of synaptophysin but also exacerbated neurological function deficits. Microassay analysis showed that 14 genes such as Wnt and Gsk3-β were downregulated after BDNF knockdown. The present data therefore showed that BDNF-mediated neuroplasticity underlie the mechanism of NSC transplantation for the treatment of TBI in adult rats.
Collapse
|
16
|
Doulames VM, Plant GW. Induced Pluripotent Stem Cell Therapies for Cervical Spinal Cord Injury. Int J Mol Sci 2016; 17:530. [PMID: 27070598 PMCID: PMC4848986 DOI: 10.3390/ijms17040530] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/17/2016] [Accepted: 03/28/2016] [Indexed: 02/07/2023] Open
Abstract
Cervical-level injuries account for the majority of presented spinal cord injuries (SCIs) to date. Despite the increase in survival rates due to emergency medicine improvements, overall quality of life remains poor, with patients facing variable deficits in respiratory and motor function. Therapies aiming to ameliorate symptoms and restore function, even partially, are urgently needed. Current therapeutic avenues in SCI seek to increase regenerative capacities through trophic and immunomodulatory factors, provide scaffolding to bridge the lesion site and promote regeneration of native axons, and to replace SCI-lost neurons and glia via intraspinal transplantation. Induced pluripotent stem cells (iPSCs) are a clinically viable means to accomplish this; they have no major ethical barriers, sources can be patient-matched and collected using non-invasive methods. In addition, the patient’s own cells can be used to establish a starter population capable of producing multiple cell types. To date, there is only a limited pool of research examining iPSC-derived transplants in SCI—even less research that is specific to cervical injury. The purpose of the review herein is to explore both preclinical and clinical recent advances in iPSC therapies with a detailed focus on cervical spinal cord injury.
Collapse
Affiliation(s)
- Vanessa M Doulames
- Stanford Partnership for Spinal Cord Injury and Repair, Department of Neurosurgery, Stanford University School of Medicine, 265 Campus Drive Stanford, California, CA 94305, USA.
| | - Giles W Plant
- Stanford Partnership for Spinal Cord Injury and Repair, Department of Neurosurgery, Stanford University School of Medicine, 265 Campus Drive Stanford, California, CA 94305, USA.
| |
Collapse
|
17
|
Kumar S, Babiarz J, Basak S, Kim JH, Barminko J, Gray A, Mendapara P, Schloss R, Yarmush ML, Grumet M. Sizes and Sufficient Quantities of MSC Microspheres for Intrathecal Injection to Modulate Inflammation in Spinal Cord Injury. ACTA ACUST UNITED AC 2016; 5. [PMID: 29545904 DOI: 10.1142/s179398441550004x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Microencapsulation of mesenchymal stem cells (MSC) in alginate facilitates cell delivery, localization and survival, and modulates inflammation in vivo. However, we found that delivery of the widely used ~0.5 mm diameter encapsulated MSC (eMSC) by intrathecal injection into spinal cord injury (SCI) rats was highly variable. Injections of smaller (~0.2 mm) diameter eMSC into the lumbar spine were much more reproducible and they increased the anti-inflammatory macrophage response around the SCI site. We now report that injection of small eMSC >2 cm caudal from the rat SCI improved locomotion and myelin preservation 8 weeks after rat SCI versus control injections. Because preparation of sufficient quantities of small eMSC for larger studies was not feasible and injection of the large eMSC is problematic, we have developed a procedure to prepare medium-sized eMSC (~0.35 mm diameter) that can be delivered more reproducibly into the lumbar rat spine. The number of MSC incorporated/capsule in the medium sized capsules was ~5-fold greater than that in small capsules and the total yield of eMSC was ~20-fold higher than that for the small capsules. Assays with all three sizes of eMSC capsules showed that they inhibited TNF-α secretion from activated macrophages in co-cultures, suggesting no major difference in their anti-inflammatory activity in vitro. The in vivo activity of the medium-sized eMSC was tested after injecting them into the lumbar spine 1 day after SCI. Histological analyses 1 week later showed that eMSC reduced levels of activated macrophages measured by IB4 staining and increased white matter sparing in similar regions adjacent to the SCI site. The combined results indicate that ~0.35 mm diameter eMSC reduced macrophage inflammation in regions where white matter was preserved during critical early phases after SCI. These techniques enable preparation of eMSC in sufficient quantities to perform pre-clinical SCI studies with much larger numbers of subjects that will provide functional analyses of several critical parameters in rodent models for CNS inflammatory injury.
Collapse
Affiliation(s)
- Suneel Kumar
- Department of Cell Biology & Neuroscience, Rutgers University, 604 Allison Rd., Piscataway, NJ 08854 USA
| | - Joanne Babiarz
- Department of Cell Biology & Neuroscience, Rutgers University, 604 Allison Rd., Piscataway, NJ 08854 USA
| | - Sayantani Basak
- Department of Cell Biology & Neuroscience, Rutgers University, 604 Allison Rd., Piscataway, NJ 08854 USA
| | - Jae Hwan Kim
- Department of Cell Biology & Neuroscience, Rutgers University, 604 Allison Rd., Piscataway, NJ 08854 USA. Department of Anatomy, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Jeffrey Barminko
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854 USA. The Mount Sinai Hospital, One Gustave L. Levy Place New York, NY 10029
| | - Andrea Gray
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854 USA
| | - Parry Mendapara
- Department of Cell Biology & Neuroscience, Rutgers University, 604 Allison Rd., Piscataway, NJ 08854 USA
| | - Rene Schloss
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854 USA
| | - Martin L Yarmush
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854 USA
| | - Martin Grumet
- W. M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center. Department of Cell Biology & Neuroscience, Rutgers University, Piscataway, NJ, 08854 USA
| |
Collapse
|
18
|
Lukovic D, Stojkovic M, Moreno-Manzano V, Jendelova P, Sykova E, Bhattacharya SS, Erceg S. Concise review: reactive astrocytes and stem cells in spinal cord injury: good guys or bad guys? Stem Cells 2016; 33:1036-41. [PMID: 25728093 DOI: 10.1002/stem.1959] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 12/02/2014] [Accepted: 12/07/2014] [Indexed: 12/26/2022]
Abstract
Spinal cord injury (SCI) usually results in long lasting locomotor and sensory neuron degeneration below the injury. Astrocytes normally play a decisive role in mechanical and metabolic support of neurons, but in the spinal cord they cause injury, exerting well-known detrimental effects that contribute to glial scar formation and inhibition of axon outgrowth. Cell transplantation is considered a promising approach for replacing damaged cells and promoting neuroprotective and neuroregenerative repair, but the effects of the grafted cells on local tissue and the regenerative properties of endogenous neural stem cells in the injured spinal cord are largely unknown. During the last 2 decades cumulative evidence from diverse animal models has indicated that reactive astrocytes in synergy with transplanted cells could be beneficial for injury in multiple ways, including neuroprotection and axonal growth. In this review, we specifically focus on the dual opposing roles of reactive astrocytes in SCI and how they contribute to the creation of a permissive environment when combined with transplanted cells as the influential components for a local regenerative niche. Modulation of reactive astrocyte function might represent an extremely attractive new therapy to enhance the functional outcomes in patients.
Collapse
Affiliation(s)
- Dunja Lukovic
- CABIMER (Centro Andaluz de Biología Molecular y Medicina Regenerativa), Avda. Americo Vespucio s/n, Parque Científico y Tecnológico Cartuja, Sevilla, Spain
| | | | | | | | | | | | | |
Collapse
|
19
|
White RE, Barry DS. The emerging roles of transplanted radial glial cells in regenerating the central nervous system. Neural Regen Res 2015; 10:1548-51. [PMID: 26692835 PMCID: PMC4660731 DOI: 10.4103/1673-5374.165317] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Robin E White
- Biology Department, Westfield State University, Westfield, MA, USA
| | - Denis S Barry
- Department of Anatomy, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Ireland
| |
Collapse
|
20
|
Mashanov VS, Zueva OR, García-Arrarás JE. Heterogeneous generation of new cells in the adult echinoderm nervous system. Front Neuroanat 2015; 9:123. [PMID: 26441553 PMCID: PMC4585025 DOI: 10.3389/fnana.2015.00123] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/29/2015] [Indexed: 11/13/2022] Open
Abstract
Adult neurogenesis, generation of new functional cells in the mature central nervous system (CNS), has been documented in a number of diverse organisms, ranging from humans to invertebrates. However, the origin and evolution of this phenomenon is still poorly understood for many of the key phylogenetic groups. Echinoderms are one such phylum, positioned as a sister group to chordates within the monophyletic clade Deuterostomia. They are well known for the ability of their adult organs, including the CNS, to completely regenerate after injury. Nothing is known, however, about production of new cells in the nervous tissue under normal physiological conditions in these animals. In this study, we show that new cells are continuously generated in the mature radial nerve cord (RNC) of the sea cucumber Holothuria glaberrima. Importantly, this neurogenic activity is not evenly distributed, but is significantly more extensive in the lateral regions of the RNC than along the midline. Some of the new cells generated in the apical region of the ectoneural neuroepithelium leave their place of origin and migrate basally to populate the neural parenchyma. Gene expression analysis showed that generation of new cells in the adult sea cucumber CNS is associated with transcriptional activity of genes known to be involved in regulation of various aspects of neurogenesis in other animals. Further analysis of one of those genes, the transcription factor Myc, showed that it is expressed, in some, but not all radial glial cells, suggesting heterogeneity of this CNS progenitor cell population in echinoderms.
Collapse
Affiliation(s)
| | - Olga R Zueva
- Department of Biology, University of Puerto Rico Rio Piedras, PR, USA
| | | |
Collapse
|
21
|
Zhang S, Burda JE, Anderson MA, Zhao Z, Ao Y, Cheng Y, Sun Y, Deming TJ, Sofroniew MV. Thermoresponsive Copolypeptide Hydrogel Vehicles for Central Nervous System Cell Delivery. ACS Biomater Sci Eng 2015; 1:705-717. [PMID: 27547820 PMCID: PMC4991036 DOI: 10.1021/acsbiomaterials.5b00153] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Biomaterial vehicles have the potential to facilitate cell transplantation in the central nervous system (CNS). We have previously shown that highly tunable ionic diblock copolypeptide hydrogels (DCH) can provide sustained release of hydrophilic and hydrophobic molecules in the CNS. Here, we show that recently developed non-ionic and thermoresponsive DCH called DCHT exhibit excellent cytocompatibility. Neural stem cell (NSC) suspensions in DCHT were easily injected as liquids at room temperature. DCHT with a viscosity tuned to prevent cell sedimentation and clumping significantly increased the survival of NSC passed through injection cannulae. At body temperature, DCHT self-assembled into hydrogels with a stiffness tuned to that of CNS tissue. After injection in vivo, DCHT significantly increased by three-fold the survival of NSC grafted into healthy CNS. In injured CNS, NSC injected as suspensions in DCHT distributed well in non-neural lesion cores, integrated with healthy neural cells at lesion perimeters and supported regrowing host nerve fibers. Our findings show that non-ionic DCHT have numerous advantageous properties that make them useful tools for in vivo delivery of cells and molecules in the CNS for experimental investigations and potential therapeutic strategies.
Collapse
Affiliation(s)
- Shanshan Zhang
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles CA 90095-1569, USA
| | - Joshua E. Burda
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA 90095-1763, USA
| | - Mark A. Anderson
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA 90095-1763, USA
| | - Ziru Zhao
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA 90095-1763, USA
| | - Yan Ao
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA 90095-1763, USA
| | - Yin Cheng
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA 90095-1763, USA
| | - Yi Sun
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA 90095-1763, USA
| | - Timothy J. Deming
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles CA 90095-1569, USA
- Department of Bioengineering, University of California Los Angeles, Los Angeles CA 90095-1600, USA
| | - Michael V. Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles CA 90095-1763, USA
| |
Collapse
|
22
|
Myc regulates programmed cell death and radial glia dedifferentiation after neural injury in an echinoderm. BMC DEVELOPMENTAL BIOLOGY 2015; 15:24. [PMID: 26025619 PMCID: PMC4448152 DOI: 10.1186/s12861-015-0071-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 05/01/2015] [Indexed: 11/30/2022]
Abstract
Background Adult echinoderms can completely regenerate major parts of their central nervous system even after severe injuries. Even though this capacity has long been known, the molecular mechanisms that drive fast and complete regeneration in these animals have remained uninvestigated. The major obstacle for understanding these molecular pathways has been the lack of functional genomic studies on regenerating adult echinoderms. Results Here, we employ RNA interference-mediated gene knockdown to characterize the role of Myc during the early (first 48 hours) post-injury response in the radial nerve cord of the sea cucumber Holothuria glaberrima. Our previous experiments identified Myc as the only pluripotency-associated factor, whose expression significantly increased in the wounded CNS. The specific function(s) of this gene, however, remained unknown. Here we demonstrate that knockdown of Myc inhibits dedifferentiation of radial glia and programmed cell death, the two most prominent cellular events that take place in the regenerating sea cucumber nervous system shortly after injury. Conclusions In this study, we show that Myc overexpression is required for proper dedifferentiation of radial glial cells and for triggering the programmed cell death in the vicinity of the injury. Myc is thus the first transcription factor, whose functional role has been experimentally established in echinoderm regeneration. Electronic supplementary material The online version of this article (doi:10.1186/s12861-015-0071-z) contains supplementary material, which is available to authorized users.
Collapse
|
23
|
Chemokine-ligands/receptors: multiplayers in traumatic spinal cord injury. Mediators Inflamm 2015; 2015:486758. [PMID: 25977600 PMCID: PMC4419224 DOI: 10.1155/2015/486758] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/23/2015] [Indexed: 12/29/2022] Open
Abstract
Spinal cord injury (SCI) results in complex posttraumatic sequelae affecting the whole neuraxis. Due to its involvement in varied neuromodulatory processes, the chemokine-ligand/receptor-network is a key element of secondary lesion cascades induced by SCI. This review will provide a synopsis of chemokine-ligand/receptor-expression along the whole neuraxis after traumatic spinal cord (sc) insults on basis of recent in vivo and in vitro findings in a SCI paradigm of thoracic force-defined impact lesions (Infinite Horizon Impactor) in adult rats. Analyses of chemokine-ligand/receptor-expression at defined time points after sc lesion of different severity grades or sham operation revealed that these inflammatory mediators are induced in distinct anatomical sc regions and in thalamic nuclei, periaqueductal grey, and hippocampal structures in the brain. Cellular and anatomical expression profiles together with colocalization/expression of neural stem/progenitor cell markers in adult sc stem cells niches or with pain-related receptors and mediators in dorsal horns, dorsal columns, and pain-processing brain areas support the notion that chemokines are involved in distinct cascades underlying clinical posttraumatic impairments and syndromes. These aspects and their implication in concepts of tailored SCI treatment are reviewed in the context of the recent literature on chemokine-ligand/receptor involvement in complex secondary lesion cascades.
Collapse
|
24
|
Mashanov VS, Zueva OR, García-Arrarás JE. Expression of pluripotency factors in echinoderm regeneration. Cell Tissue Res 2014; 359:521-536. [PMID: 25468557 DOI: 10.1007/s00441-014-2040-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 10/16/2014] [Indexed: 12/15/2022]
Abstract
Cell dedifferentiation is an integral component of post-traumatic regeneration in echinoderms. As dedifferentiated cells become multipotent, we asked if this spontaneous broadening of developmental potential is associated with the action of the same pluripotency factors (known as Yamanaka factors) that were used to induce pluripotency in specialized mammalian cells. In this study, we investigate the expression of orthologs of the four Yamanaka factors in regeneration of two different organs, the radial nerve cord and the digestive tube, in the sea cucumber Holothuria glaberrima. All four pluripotency factors are expressed in uninjured animals, although their expression domains do not always overlap. In regeneration, the expression levels of the four genes were not regulated in a coordinated way, but instead showed different dynamics for individual genes and also were different between the radial nerve and the gut. SoxB1, the ortholog of the mammalian Sox2, was drastically downregulated in the regenerating intestine, suggesting that this factor is not required for dedifferentiation/regeneration in this organ. On the other hand, during the early post-injury stage, Myc, the sea cucumber ortholog of c-Myc, was significantly upregulated in both the intestine and the radial nerve cord and is therefore hypothesized to play a central role in dedifferentiation/regeneration of various tissue types.
Collapse
Affiliation(s)
- Vladimir S Mashanov
- Department of Biology, University of Puerto Rico, PO Box 70377, San Juan, PR, 00936-8377, USA.
| | - Olga R Zueva
- Department of Biology, University of Puerto Rico, PO Box 70377, San Juan, PR, 00936-8377, USA
| | - José E García-Arrarás
- Department of Biology, University of Puerto Rico, PO Box 70377, San Juan, PR, 00936-8377, USA
| |
Collapse
|
25
|
Iwai H, Nori S, Nishimura S, Yasuda A, Takano M, Tsuji O, Fujiyoshi K, Toyama Y, Okano H, Nakamura M. Transplantation of Neural Stem/Progenitor Cells at Different Locations in Mice with Spinal Cord Injury. Cell Transplant 2014; 23:1451-64. [DOI: 10.3727/096368913x670967] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transplantation of neural stem/progenitor cells (NS/PCs) promotes functional recovery after spinal cord injury (SCI); however, few studies have examined the optimal site of NS/PC transplantation in the spinal cord. The purpose of this study was to determine the optimal transplantation site of NS/PCs for the treatment of SCI. Wild-type mice were generated with contusive SCI at the T10 level, and NS/PCs were derived from fetal transgenic mice. These NS/PCs ubiquitously expressed ffLuc-cp156 protein (Venus and luciferase fusion protein) and so could be detected by in vivo bioluminescence imaging 9 days postinjury. NS/PCs (low: 250,000 cells per mouse; high: 1 million cells per mouse) were grafted into the spinal cord at the lesion epicenter (E) or at rostral and caudal (RC) sites. Phosphate-buffered saline was injected into E as a control. Motor functional recovery was better in each of the transplantation groups (E-Low, E-High, RC-Low, and RC-High) than in the control group. The photon counts of the grafted NS/PCs were similar in each of the four transplantation groups, suggesting that the survival of NS/PCs was fairly uniform when more than a certain threshold number of cells were transplanted. Quantitative RT-PCR analyses demonstrated that brain-derived neurotropic factor expression was higher in the RC segment than in the E segment, and this may underlie why NS/PCs more readily differentiated into neurons than into astrocytes in the RC group. The location of the transplantation site did not affect the area of spared fibers, angiogenesis, or the expression of any other mediators. These findings indicated that the microenvironments of the E and RC sites are able to support NS/PCs transplanted during the subacute phase of SCI similarly. Optimally, a certain threshold number of NS/PCs should be grafted into the E segment to avoid damaging sites adjacent to the lesion during the injection procedure.
Collapse
Affiliation(s)
- Hiroki Iwai
- Department of Orthopaedic Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Satoshi Nori
- Department of Orthopaedic Surgery, Tokyo Dental College Ichikawa General Hospital, Ichikawa, Chiba, Japan
| | - Soraya Nishimura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Akimasa Yasuda
- Department of Orthopaedic Surgery, National Hospital Organization, Murayama Medical Center, Musashimurayama, Tokyo, Japan
| | - Morito Takano
- Department of Orthopaedic Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Osahiko Tsuji
- Department of Orthopaedic Surgery, Saitama Social Insurance Hospital, Urawa, Saitama, Japan
| | - Kanehiro Fujiyoshi
- Department of Orthopaedic Surgery, National Hospital Organization, Murayama Medical Center, Musashimurayama, Tokyo, Japan
| | - Yoshiaki Toyama
- Department of Orthopaedic Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Masaya Nakamura
- Department of Orthopaedic Surgery, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| |
Collapse
|
26
|
Jin L, Wu Z, Xu W, Hu X, Zhang J, Xue Z, Cheng L. Identifying gene expression profile of spinal cord injury in rat by bioinformatics strategy. Mol Biol Rep 2014; 41:3169-77. [PMID: 24595446 DOI: 10.1007/s11033-014-3176-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 01/16/2014] [Indexed: 01/12/2023]
Abstract
Spinal cord injury (SCI) leads to the loss of sensory, motor, and autonomic function. We aimed to identify the therapeutic targets of-SCI by bioinformatics analysis. The gene expression profile of GSE20907 was downloaded from gene expression omnibus database. By comparing gene expression profiles with control samples, we screened out several differentially expressed genes (DEGs) in 3 days, 2 weeks and 1 month post-SCI. The pathway enrichment and protein-protein interaction (PPI) network analysis for the identified DEGs were performed. Then, transcription factors and microRNAs for DEGs were predicted. We found that up-regulated DEGs mainly participated in cell cycle, oxidative phosphorylation and immune-related pathways; while down-regulated DEGs were mainly involved in oxidative phosphorylation and central nervous system disease signaling pathways. In the constructed PPI network, Bub1, Vascular endothelial growth factor, Topoisomerase IIα (TOP2a) and Cdc20 showed better correspondence with cell cycle, repair system and nerve system. Furthermore, the up-regulated genes (Arpc1b, CD74 and Brd2) significantly mapped to the target genes of transcription factors. The down-regulated genes of 3 days post-injury and the up-regulated genes of 2 weeks post-injury were significantly enriched as the target genes of microRNAs (miR-129 and miR-124). In conclusion, our results may provide guidelines to discuss the collaboration of PPI network in carcinogenesis of SCI.
Collapse
Affiliation(s)
- Lingjing Jin
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | | | | | | | | | | | | |
Collapse
|
27
|
Goldshmit Y, Frisca F, Pinto AR, Pébay A, Tang JKKY, Siegel AL, Kaslin J, Currie PD. Fgf2 improves functional recovery-decreasing gliosis and increasing radial glia and neural progenitor cells after spinal cord injury. Brain Behav 2014; 4:187-200. [PMID: 24683512 PMCID: PMC3967535 DOI: 10.1002/brb3.172] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Accepted: 07/30/2013] [Indexed: 01/26/2023] Open
Abstract
OBJECTIVES A major impediment for recovery after mammalian spinal cord injury (SCI) is the glial scar formed by proliferating reactive astrocytes. Finding factors that may reduce glial scarring, increase neuronal survival, and promote neurite outgrowth are of major importance for improving the outcome after SCI. Exogenous fibroblast growth factor (Fgf) has been shown to decrease injury volume and improve functional outcome; however, the mechanisms by which this is mediated are still largely unknown. METHODS In this study, Fgf2 was administered for 2 weeks in mice subcutaneously, starting 30 min after spinal cord hemisection. RESULTS Fgf2 treatment decreased the expression of TNF-a at the lesion site, decreased monocyte/macrophage infiltration, and decreased gliosis. Fgf2 induced astrocytes to adopt a polarized morphology and increased expression of radial markers such as Pax6 and nestin. In addition, the levels of chondroitin sulfate proteoglycans (CSPGs), expressed by glia, were markedly decreased. Furthermore, Fgf2 treatment promotes the formation of parallel glial processes, "bridges," at the lesion site that enable regenerating axons through the injury site. Additionally, Fgf2 treatment increased Sox2-expressing cells in the gray matter and neurogenesis around and at the lesion site. Importantly, these effects were correlated with enhanced functional recovery of the left paretic hind limb. CONCLUSIONS Thus, early pharmacological intervention with Fgf2 following SCI is neuroprotective and creates a proregenerative environment by the modulation of the glia response.
Collapse
Affiliation(s)
- Yona Goldshmit
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia ; Centre for Eye Research Australia & Royal Victorian Eye and Ear Hospital East Melbourne, VIC, Australia
| | - Frisca Frisca
- Department of Ophthalmology, The University of Melbourne East Melbourne, VIC, Australia
| | - Alexander R Pinto
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia
| | - Alice Pébay
- Centre for Eye Research Australia & Royal Victorian Eye and Ear Hospital East Melbourne, VIC, Australia ; Department of Ophthalmology, The University of Melbourne East Melbourne, VIC, Australia
| | | | - Ashley L Siegel
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia
| | - Jan Kaslin
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia
| | - Peter D Currie
- Australian Regenerative Medicine Institute East Melbourne, VIC, Australia
| |
Collapse
|
28
|
Skop NB, Calderon F, Cho CH, Gandhi CD, Levison SW. Optimizing a multifunctional microsphere scaffold to improve neural precursor cell transplantation for traumatic brain injury repair. J Tissue Eng Regen Med 2013; 10:E419-E432. [DOI: 10.1002/term.1832] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 06/12/2013] [Accepted: 09/02/2013] [Indexed: 12/16/2022]
Affiliation(s)
- Nolan B. Skop
- Department of Neurology and Neurosciences; Rutgers-New Jersey Medical School; Newark NJ USA
- Department of Neurological Surgery; Rutgers-New Jersey Medical School; Newark NJ USA
| | - Frances Calderon
- Department of Neurology and Neurosciences; Rutgers-New Jersey Medical School; Newark NJ USA
| | - Cheul H. Cho
- Department of Biomedical Engineering; New Jersey Institute of Technology; Newark NJ USA
| | - Chirag D. Gandhi
- Department of Neurological Surgery; Rutgers-New Jersey Medical School; Newark NJ USA
| | - Steven W. Levison
- Department of Neurology and Neurosciences; Rutgers-New Jersey Medical School; Newark NJ USA
| |
Collapse
|
29
|
Xia GN, Zou Y, Wang YC, Xia QJ, Lu BT, Wang TH, Qi JG. Neural stem cells grafts decrease neural apoptosis associated with caspase-7 downregulation and BDNF upregulation in rats following spinal cord hemisection. Cell Mol Neurobiol 2013; 33:1013-22. [PMID: 23963709 PMCID: PMC11497902 DOI: 10.1007/s10571-013-9969-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 07/23/2013] [Indexed: 12/11/2022]
Abstract
Transplantation of neural stem cells (NSCs) into lesioned spinal cord demonstrated a beneficial effect for neural repair, the underlying mechanism, however, remains to be elusive. Here, we showed that NSCs, possessing the capacity to differentiate toward into neurons and astrocytes, exhibit a neuroprotective effect by anti-apoptosis mechanism in spinal cord hemi-transected rats despite it did not improve behavior. Intravenous NSCs injection substantially upregulated the level of BDNF mRNA but not its receptor TrkB in hemisected spinal cord, while caspase-7, a downstream apoptosis gene of caspase-3, has been largely down-regulated. TUNEL staining showed that the number of apoptosis cells in injured spinal cord decreased significantly, compared with seen in rats with no NSCs administration. The present finding therefore provided crucial evidence to explain neuroprotective effect of NSCs grafts in hemisected spinal cord, which is associated with BDNF upregulation and caspase-7 downregulation.
Collapse
Affiliation(s)
- Guan-nan Xia
- Department of Histology, Embryology and Neurobiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, 610041 China
| | - Yu Zou
- Institute of Neuroscience, Kunming Medical University, Kunming, 650031 China
| | - You-cui Wang
- Department of Histology, Embryology and Neurobiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, 610041 China
| | - Qing-jie Xia
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Bing-tuan Lu
- Institute of Neuroscience, Kunming Medical University, Kunming, 650031 China
| | - Ting-hua Wang
- Institute of Neuroscience, Kunming Medical University, Kunming, 650031 China
- Institute of Neurological Disease, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Jian-guo Qi
- Department of Histology, Embryology and Neurobiology, West China School of Preclinical and Forensic Medicine, Sichuan University, Chengdu, 610041 China
| |
Collapse
|
30
|
Abstract
Oxidative stress is a widely recognized cause of cell death associated with neurodegeneration, inflammation, and aging. Tyrosine nitration in these conditions has been reported extensively, but whether tyrosine nitration is a marker or plays a role in the cell-death processes was unknown. Here, we show that nitration of a single tyrosine residue on a small proportion of 90-kDa heat-shock protein (Hsp90), is sufficient to induce motor neuron death by the P2X7 receptor-dependent activation of the Fas pathway. Nitrotyrosine at position 33 or 56 stimulates a toxic gain of function that turns Hsp90 into a toxic protein. Using an antibody that recognizes the nitrated Hsp90, we found immunoreactivity in motor neurons of patients with amyotrophic lateral sclerosis, in an animal model of amyotrophic lateral sclerosis, and after experimental spinal cord injury. Our findings reveal that cell death can be triggered by nitration of a single protein and highlight nitrated Hsp90 as a potential target for the development of effective therapies for a large number of pathologies.
Collapse
|
31
|
Barry DS, Pakan JMP, O'Keeffe GW, McDermott KW. The spatial and temporal arrangement of the radial glial scaffold suggests a role in axon tract formation in the developing spinal cord. J Anat 2012; 222:203-13. [PMID: 23121514 DOI: 10.1111/joa.12006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2012] [Indexed: 01/14/2023] Open
Abstract
Radial glial cells serve diverse roles during the development of the central nervous system (CNS). In the embryonic brain, they are recognised as guidance conduits for migrating neuroblasts and as multipotent stem cells, generating both neurons and glia. While their stem cell capacities in the developing spinal cord are as yet not fully clarified, they are classically seen as a population of astrocytes precursors, before gradually disappearing as the spinal cord matures. Although the origins and lineages of CNS radial glial cells are being more clearly understood, the relationships between radial glial cells and growing white matter (WM) tracts are largely unknown. Here, we provide an in-depth description of the distribution and organisation of radial glial cell processes during the peak periods of axonogenesis in the rat spinal cord. We show that radial glial cell distribution is highly ordered in the WM from E14 to E18, when the initial patterning of axon tracts is taking place. We report that the density of radial glial cell processes is tightly conserved throughout development in the dorsal, lateral and ventral WM funiculi along the rostrocaudal axis of the spinal cord. We provide evidence that from E16 the dorsal funiculi grow within and are segregated by fascicles of processes emanating from the dorsomedial septum. The density of radial glial cells declines with the maturation of axon tracts and coincides with the onset of the radial glial cell-astrocyte transformation. As such, we propose that radial glial cells act as structural scaffolds by compartmentalising and supporting WM patterning in the spinal cord during embryonic development.
Collapse
Affiliation(s)
- Denis S Barry
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | | | | | | |
Collapse
|
32
|
Roach P, Parker T, Gadegaard N, Alexander MR. A bio-inspired neural environment to control neurons comprising radial glia, substrate chemistry and topography. Biomater Sci 2012; 1:83-93. [PMID: 32481998 DOI: 10.1039/c2bm00060a] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Achieving alignment of cells is key to the success of regenerative strategies of neural tissue. We report a high-throughput method to investigate neural cell response to surface chemistry overlaid orthogonally onto a gradient of gradually changing groove widths. Using a bio-inspired approach wherein radial glial cells, which naturally guide neurons in the developing brain, enhance the attachment and directional outgrowth of neurons, we show the differences in the interaction and cellular response of glia, neurons and co-cultured cells. Radial glia were found to preferentially reside in grooves of width 6-35 μm with greater alignment to grooves <10 μm on the hydrophobic and hydrophilic extremes of chemistry. When neurons were sequentially cultured after radial glia, they showed enhanced alignment compared to when they were cultured alone, for all chemistries and groove widths. This is not dependent on co-localisation of the neurons with glia suggesting the radial glial cells pre-condition the substrate giving rise to enhanced attachment and alignment of subsequently cultured neurons. The results indicate a dependence of both primary radial glia and neuron responses on surface chemistry and micro-groove width. Grooved surfaces (width 5-10 μm) of mid-range wettability show the greatest potential to significantly enhance axonal alignment and, therefore, potential regeneration, when pre-conditioned by radial glia, highlighting the importance of surface engineering for neural scaffolds.
Collapse
Affiliation(s)
- Paul Roach
- Laboratory of Biophysics and Surface Analysis, School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | | | | | | |
Collapse
|
33
|
Li H, Hader AT, Han YR, Wong JA, Babiarz J, Ricupero CL, Godfrey SB, Corradi JP, Fennell M, Hart RP, Plummer MR, Grumet M. Isolation of a novel rat neural progenitor clone that expresses Dlx family transcription factors and gives rise to functional GABAergic neurons in culture. Dev Neurobiol 2012; 72:805-20. [PMID: 21913335 DOI: 10.1002/dneu.20977] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Gamma-aminobutyric acid (GABA) ergic interneurons are lost in conditions including epilepsy and central nervous system injury, but there are few culture models available to study their function. Toward the goal of obtaining renewable sources of GABAergic neurons, we used the molecular profile of a functionally incomplete GABAergic precursor clone to screen 17 new clones isolated from GFP(+) rat E14.5 cortex and ganglionic eminence (GE) that were generated by viral introduction of v-myc. The clones grow as neurospheres in medium with FGF2, and after withdrawal of FGF2, they exhibit varying patterns of differentiation. Transcriptional profiling and quantitative reverse transcriptase polymerase chain reaction (RT-PCR) indicated that one clone (GE6) expresses high levels of mRNAs encoding Dlx1, 2, 5, and 6, glutamate decarboxylases, and presynaptic proteins including neuropeptide Y and somatostatin. Protein expression confirmed that GE6 is a progenitor with restricted differentiation giving rise mostly to neurons with GABAergic markers. In cocultures with hippocampal neurons, GE6 neurons became electrically excitable and received both inhibitory and excitatory synapses. After withdrawal of FGF2 in cultures of GE6 alone, neurons matured to express βIII-tubulin, and staining for synaptophysin and vesicular GABA transporter were robust after 1-2 weeks of differentiation. GE6 neurons also became electrically excitable and displayed synaptic activity, but synaptic currents were carried by chloride and were blocked by bicuculline. The results suggest that the GE6 clone, which is ventrally derived from the GE, resembles GABAergic interneuron progenitors that migrate into the developing forebrain. This is the first report of a relatively stable fetal clone that can be differentiated into GABAergic interneurons with functional synapses.
Collapse
Affiliation(s)
- Hedong Li
- W.M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Rutgers, State University of New Jersey, Piscataway, New Jersey 08854-8082, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Adult zebrafish show a remarkable capacity to regenerate their spinal column after injury, an ability that stands in stark contrast to the limited repair that occurs within the mammalian CNS post-injury. The reasons for this interspecies difference in regenerative capacity remain unclear. Here we demonstrate a novel role for Fgf signaling during glial cell morphogenesis in promoting axonal regeneration after spinal cord injury. Zebrafish glia are induced by Fgf signaling, to form an elongated bipolar morphology that forms a bridge between the two sides of the resected spinal cord, over which regenerating axons actively migrate. Loss of Fgf function inhibits formation of this "glial bridge" and prevents axon regeneration. Despite the poor potential for mammalian axonal regeneration, primate astrocytes activated by Fgf signaling adopt a similar morphology to that induced in zebrafish glia. This suggests that differential Fgf regulation, rather than intrinsic cell differences, underlie the distinct responses of mammalian and zebrafish glia to injury.
Collapse
|
35
|
Otsuka S, Adamson C, Sankar V, Gibbs KM, Kane-Goldsmith N, Ayer J, Babiarz J, Kalinski H, Ashush H, Alpert E, Lahav R, Feinstein E, Grumet M. Delayed intrathecal delivery of RhoA siRNA to the contused spinal cord inhibits allodynia, preserves white matter, and increases serotonergic fiber growth. J Neurotrauma 2012; 28:1063-76. [PMID: 21443453 DOI: 10.1089/neu.2010.1568] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
RhoA is a key regulator of the actin cytoskeleton that is upregulated after spinal cord injury (SCI). We analyzed different methods for siRNA delivery and developed siRNAs targeting RhoA (siRhoA) for SCI treatment. Cy 3.5-labeled siRNA delivered at the time of SCI yielded fluorescence in several cell types in the injury site. Intraspinal injections of chemically stabilized siRhoA into the spinal cord of injured rats reduced RhoA protein levels after 1 week and improved hindlimb walking over 6 weeks. To explore a less invasive route, we tested intrathecal injection of Cy 3.5-labeled siRNA via lumbar puncture 1 day after SCI, which resulted in robust uptake in the T9-T10 injury site. Lumbar injection of siRhoA 1 day after SCI reduced RhoA mRNA and protein levels 3 days after injection. Although siRhoA treatment did not yield significant improvement in locomotion, it decreased tactile hypersensitivity significantly compared to controls. Histological analysis at 8 weeks showed significant improvement in white matter sparing with siRhoA compared to control siRNA. siRhoA treatment also resulted in less accumulation of ED1+macrophages, increased PKC-γ immunoreactivity in the corticospinal tract rostral to the injury site, and increased serotonergic fiber growth 12 mm caudal to the contusion site. The ability of siRhoA to preserve white matter and promote serotonergic axonal regrowth caudal to the injury site is likely to suppress allodynia. This provides justification for considering clinical development of RhoA inhibitors to treat SCI sub-acutely to reduce allodynia, which occurs frequently in SCI patients.
Collapse
Affiliation(s)
- Seiji Otsuka
- W.M. Keck Center for Collaborative Neuroscience, Rutgers Stem Cell Research Center, Department of Cell Biology & Neuroscience, Rutgers, State University of New Jersey, Piscataway, New Jersey 08854-8082, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Fawcett JW, Schwab ME, Montani L, Brazda N, Müller HW. Defeating inhibition of regeneration by scar and myelin components. HANDBOOK OF CLINICAL NEUROLOGY 2012; 109:503-22. [PMID: 23098733 DOI: 10.1016/b978-0-444-52137-8.00031-0] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Axon regeneration and the sprouting processes that underlie plasticity are blocked by inhibitory factors in the central nervous system (CNS) environment, several of which are upregulated after injury. The major inhibitory molecules are those associated with myelin and those associated with the glial scar. In myelin, NogoA, MAG, and OMgp are present on normal oligodendrocytes and on myelin debris. They act partly via the Nogo receptor, partly via an unidentified amino-Nogo receptor. In the glial scar, chondroitin sulphate proteoglycans, semaphorins, and the formation of a collagen-based membrane are all inhibitory. Methods to counteract these forms of inhibition have been identified, and these treatments promote axon regeneration in the damaged spinal cord, and in some cases recovery of function through enhanced plasticity.
Collapse
Affiliation(s)
- James W Fawcett
- Cambridge University Centre for Brain Repair, Cambridge, UK.
| | | | | | | | | |
Collapse
|
37
|
Transforming growth factor α transforms astrocytes to a growth-supportive phenotype after spinal cord injury. J Neurosci 2011; 31:15173-87. [PMID: 22016551 DOI: 10.1523/jneurosci.3441-11.2011] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Astrocytes are both detrimental and beneficial for repair and recovery after spinal cord injury (SCI). These dynamic cells are primary contributors to the growth-inhibitory glial scar, yet they are also neuroprotective and can form growth-supportive bridges on which axons traverse. We have shown that intrathecal administration of transforming growth factor α (TGFα) to the contused mouse spinal cord can enhance astrocyte infiltration and axonal growth within the injury site, but the mechanisms of these effects are not well understood. The present studies demonstrate that the epidermal growth factor receptor (EGFR) is upregulated primarily by astrocytes and glial progenitors early after SCI. TGFα directly activates the EGFR on these cells in vitro, inducing their proliferation, migration, and transformation to a phenotype that supports robust neurite outgrowth. Overexpression of TGFα in vivo by intraparenchymal adeno-associated virus injection adjacent to the injury site enhances cell proliferation, alters astrocyte distribution, and facilitates increased axonal penetration at the rostral lesion border. To determine whether endogenous EGFR activation is required after injury, SCI was also performed on Velvet (C57BL/6J-Egfr(Vel)/J) mice, a mutant strain with defective EGFR activity. The affected mice exhibited malformed glial borders, larger lesions, and impaired recovery of function, indicating that intrinsic EGFR activation is necessary for neuroprotection and normal glial scar formation after SCI. By further stimulating precursor proliferation and modifying glial activation to promote a growth-permissive environment, controlled stimulation of EGFR at the lesion border may be considered in the context of future strategies to enhance endogenous cellular repair after injury.
Collapse
|
38
|
Barminko J, Kim JH, Otsuka S, Gray A, Schloss R, Grumet M, Yarmush ML. Encapsulated mesenchymal stromal cells for in vivo transplantation. Biotechnol Bioeng 2011; 108:2747-58. [PMID: 21656712 PMCID: PMC3178737 DOI: 10.1002/bit.23233] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 05/20/2011] [Accepted: 05/23/2011] [Indexed: 12/13/2022]
Abstract
Immunomodulatory human mesenchymal stromal cells (hMSC) have been incorporated into therapeutic protocols to treat secondary inflammatory responses post-spinal cord injury (SCI) in animal models. However, limitations with direct hMSC implantation approaches may prevent effective translation for therapeutic development of hMSC infusion into post-SCI treatment protocols. To circumvent these limitations, we investigated the efficacy of alginate microencapsulation in developing an implantable vehicle for hMSC delivery. Viability and secretory function were maintained within the encapsulated hMSC population, and hMSC secreted anti-inflammatory cytokines upon induction with the pro-inflammatory factors, TNF-α and IFN-γ. Furthermore, encapsulated hMSC modulated inflammatory macrophage function both in vitro and in vivo, even in the absence of direct hMSC-macrophage cell contact and promoted the alternative M2 macrophage phenotype. In vitro, this was evident by a reduction in macrophage iNOS expression with a concomitant increase in CD206, a marker for M2 macrophages. Finally, Sprague-Dawley rat spinal cords were injured at vertebra T10 via a weight drop model (NYU model) and encapsulated hMSC were administered via lumbar puncture 24 h post-injury. Encapsulated hMSC localized primarily in the cauda equina of the spinal cord. Histological assessment of spinal cord tissue 7 days post-SCI indicated that as few as 5 × 10(4) encapsulated hMSC yielded increased numbers of CD206-expressing macrophages, consistent with our in vitro studies. The combined findings support the inclusion of immobilized hMSC in post-CNS trauma tissue protective therapy, and suggest that conversion of macrophages to the M2 subset is responsible, at least in part, for tissue protection.
Collapse
Affiliation(s)
| | - Jae Hwan Kim
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Seiji Otsuka
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA
| | - Andrea Gray
- Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Rene Schloss
- Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Martin Grumet
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, NJ, USA
| | | |
Collapse
|
39
|
Xu JC, Bernreuther C, Cui YF, Jakovcevski I, Hargus G, Xiao MF, Schachner M. Transplanted L1 expressing radial glia and astrocytes enhance recovery after spinal cord injury. J Neurotrauma 2011; 28:1921-37. [PMID: 21671795 DOI: 10.1089/neu.2011.1783] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
A major obstacle for the transplantation of neural stem cells (NSCs) into the lesioned spinal cord is their predominant astrocytic differentiation after transplantation. We took advantage of this predominant astrocytic differentiation of NSCs and expressed the paradigmatic beneficial neural cell adhesion molecule L1 in radial glial cells and reactive and nonreactive astrocytes as novel cellular vehicles to express L1 under the control of the promoter for the human glial fibrillary acidic protein (GFAP-L1 NSCs). Behavioral analysis and electrophysiological H-reflex recordings revealed that mice transplanted with GFAP-L1 NSCs showed enhanced locomotor recovery in comparison to mice injected with wild type (WT) NSCs or control mice injected with phosphate-buffered saline (PBS). This functional recovery was further accelerated in mice transplanted with L1-expressing radial glial cells that had been immunoisolated from GFAP-L1 NSCs (GFAP-L1-i cells). Morphological analysis revealed that mice grafted with GFAP-L1 NSCs exhibited increased neuronal differentiation and migration of transplanted cells, as well as increased soma size and cholinergic synaptic coverage of host motoneurons and increased numbers of endogenous catecholaminergic nerve fibers caudal to the lesion site. These findings show that L1-expressing astrocytes and radial glial cells isolated from GFAP-L1 NSC cultures represent a novel strategy for improving functional recovery after spinal cord injury, encouraging the use of the human GFAP promoter to target beneficial transgene expression in transplanted stem cells.
Collapse
Affiliation(s)
- Jin-Chong Xu
- Zentrum für Molekulare Neurobiologie Hamburg, Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
40
|
Delgado-Rivera R, Griffin J, Ricupero CL, Grumet M, Meiners S, Uhrich KE. Microscale plasma-initiated patterning of electrospun polymer scaffolds. Colloids Surf B Biointerfaces 2011; 84:591-6. [PMID: 21345656 PMCID: PMC3062666 DOI: 10.1016/j.colsurfb.2011.01.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 01/08/2011] [Accepted: 01/13/2011] [Indexed: 11/24/2022]
Abstract
Microscale plasma-initiated patterning (μPIP) is a novel micropatterning technique used to create biomolecular micropatterns on polymer surfaces. The patterning method uses a polydimethylsiloxane (PDMS) stamp to selectively protect regions of an underlying substrate from oxygen plasma treatment resulting in hydrophobic and hydrophilic regions. Preferential adsorption of the biomolecules onto either the plasma-exposed (hydrophilic) or plasma-protected (hydrophobic) regions leads to the biomolecular micropatterns. In the current work, laminin-1 was applied to an electrospun polyamide nanofibrillar matrix following plasma treatment. Radial glial clones (neural precursors) selectively adhered to these patterned matrices following the contours of proteins on the surface. This work demonstrates that textured surfaces, such as nanofibrillar scaffolds, can be micropatterned to provide external chemical cues for cellular organization.
Collapse
Affiliation(s)
- Roberto Delgado-Rivera
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, 08854
- Department of Pharmacology, Robert Wood Johnson Medical School, Piscataway, New Jersey, 08854
| | - Jeremy Griffin
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, 08854
| | - Christopher L. Ricupero
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, New Jersey, 08854
| | - Martin Grumet
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, New Jersey, 08854
| | - Sally Meiners
- Department of Pharmacology, Robert Wood Johnson Medical School, Piscataway, New Jersey, 08854
| | - Kathryn E. Uhrich
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, New Jersey, 08854
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey, 08854
| |
Collapse
|
41
|
Rehermann MI, Santiñaque FF, López-Carro B, Russo RE, Trujillo-Cenóz O. Cell proliferation and cytoarchitectural remodeling during spinal cord reconnection in the fresh-water turtle Trachemys dorbignyi. Cell Tissue Res 2011; 344:415-33. [PMID: 21574060 DOI: 10.1007/s00441-011-1173-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Accepted: 04/11/2011] [Indexed: 01/22/2023]
Abstract
In fresh-water turtles, the bridge connecting the proximal and caudal stumps of transected spinal cords consists of regenerating axons running through a glial cellular matrix. To understand the process leading to the generation of the scaffold bridging the lesion, we analyzed the mitotic activity triggered by spinal injury in animals maintained alive for 20-30 days after spinal cord transection. Flow cytometry and bromodeoxyuridine (BrdU)-labeling experiments revealed a significant increment of cycling cells around the lesion epicenter. BrdU-tagged cells maintained a close association with regenerating axons. Most dividing cells expressed the brain lipid-binding protein (BLBP). Cells with BrdU-positive nuclei expressed glial fibrillary acidic protein. As spinal cord regeneration involves dynamic cell rearrangements, we explored the ultra-structure of the bridge and found cells with the aspect of immature oligodendrocytes forming an embryonic-like microenvironment. These cells supported and ensheathed regenerating axons that were recognized by immunocytological and electron-microscopical procedures. Since functional recovery depends on proper impulse transmission, we examined the anatomical axon-glia relationships near the lesion epicenter. Computer-assisted three-dimensional models revealed helical axon-glial junctions in which the intercellular space appeared to be reduced (5-7 nm). Serial-sectioning analysis revealed that fibril-containing processes provided myelinating axon sheaths. Thus, disruption of the ependymal layer elicits mitotic activity predominantly in radial glia expressing BLBP on the lateral aspects of the ependyma. These cycling cells seem to migrate and contribute to the bridge providing the main support and sheaths for regenerating axons.
Collapse
Affiliation(s)
- María Inés Rehermann
- Departamento de Neurofisiología Celular y Molecular, Avenida Italia 3318, Montevideo, Uruguay
| | | | | | | | | |
Collapse
|
42
|
Qu Z, Sun D, Young W. Lithium promotes neural precursor cell proliferation: evidence for the involvement of the non-canonical GSK-3β-NF-AT signaling. Cell Biosci 2011; 1:18. [PMID: 21711903 PMCID: PMC3125208 DOI: 10.1186/2045-3701-1-18] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Accepted: 05/03/2011] [Indexed: 12/04/2022] Open
Abstract
Lithium, a drug that has long been used to treat bipolar disorder and some other human pathogenesis, has recently been shown to stimulate neural precursor growth. However, the involved mechanism is not clear. Here, we show that lithium induces proliferation but not survival of neural precursor cells. Mechanistic studies suggest that the effect of lithium mainly involved activation of the transcription factor NF-AT and specific induction of a subset of proliferation-related genes. While NF-AT inactivation by specific inhibition of its upstream activator calcineurin antagonized the effect of lithium on the proliferation of neural precursor cells, specific inhibition of the NF-AT inhibitor GSK-3β, similar to lithium treatment, promoted neural precursor cell proliferation. One important function of lithium appeared to increase inhibitory phosphorylation of GSK-3β, leading to GSK-3β suppression and subsequent NF-AT activation. Moreover, lithium-induced proliferation of neural precursor cells was independent of its role in inositol depletion. These findings not only provide mechanistic insights into the clinical effects of lithium, but also suggest an alternative therapeutic strategy for bipolar disorder and other neural diseases by targeting the non-canonical GSK-3β-NF-AT signaling.
Collapse
Affiliation(s)
- Zhaoxia Qu
- Department of Cell Biology and Neuroscience, W, M, Keck Center for Collaborative Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | | | | |
Collapse
|
43
|
Jin Y, Neuhuber B, Singh A, Bouyer J, Lepore A, Bonner J, Himes T, Campanelli JT, Fischer I. Transplantation of human glial restricted progenitors and derived astrocytes into a contusion model of spinal cord injury. J Neurotrauma 2011; 28:579-94. [PMID: 21222572 DOI: 10.1089/neu.2010.1626] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Transplantation of neural progenitors remains a promising therapeutic approach to spinal cord injury (SCI), but the anatomical and functional evaluation of their effects is complex, particularly when using human cells. We investigated the outcome of transplanting human glial-restricted progenitors (hGRP) and astrocytes derived from hGRP (hGDA) in spinal cord contusion with respect to cell fate and host response using athymic rats to circumvent xenograft immune issues. Nine days after injury hGRP, hGDA, or medium were injected into the lesion center and rostral and caudal to the lesion, followed by behavioral testing for 8 weeks. Both hGRP and hGDA showed robust graft survival and extensive migration. The total number of cells increased 3.5-fold for hGRP, and twofold for hGDA, indicating graft expansion, but few proliferating cells remained by 8 weeks. Grafted cells differentiated into glia, predominantly astrocytes, and few remained at progenitor state. About 80% of grafted cells around the injury were glial fibrillary acidic protein (GFAP)-positive, gradually decreasing to 40-50% at a distance of 6 mm. Conversely, there were few graft-derived oligodendrocytes at the lesion, but their numbers increased away from the injury to 30-40%. Both cell grafts reduced cyst and scar formation at the injury site compared to controls. Microglia/macrophages were present at and around the lesion area, and axons grew along the spared tissue with no differences among groups. There were no significant improvements in motor function recovery as measured by the Basso, Beattie, and Bresnahan (BBB) scale and grid tests in all experimental groups. Cystometry revealed that hGRP grafts attenuated hyperactive bladder reflexes. Importantly, there was no increased sensory or tactile sensitivity associated with pain, and the hGDA group showed sensory function returning to normal. Although the improved lesion environment was not sufficient for robust functional recovery, the permissive properties and lack of sensory hypersensitivity indicate that human GRP and astrocytes remain promising candidates for therapy after SCI.
Collapse
Affiliation(s)
- Ying Jin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jakeman LB, Hoschouer EL, Basso DM. Injured mice at the gym: review, results and considerations for combining chondroitinase and locomotor exercise to enhance recovery after spinal cord injury. Brain Res Bull 2011; 84:317-26. [PMID: 20558254 PMCID: PMC3030989 DOI: 10.1016/j.brainresbull.2010.06.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 06/02/2010] [Accepted: 06/02/2010] [Indexed: 01/08/2023]
Abstract
Exercise provides a number of important benefits after spinal cord injury in clinical studies and animal models. However, the amount of functional improvement in overground locomotion obtained with exercise alone has been limited thus far, for reasons that are still poorly understood. One hypothesis is that the complex network of endogenous extracellular matrix components, including chondroitin sulfate proteoglycans (CSPGs), can inhibit exercise-induced remodeling and limit plasticity of spared circuitry in the adult central nervous system. Recent animal studies have shown that chondroitinase ABC (ChABC) can enhance plasticity in the adult nervous system by cleaving glycosaminoglycan sidechains from CSPGs. In this article we review the current literature on plasticity observed with locomotor training and following degradation of CSPGs with ChABC and then present a rationale for the use of exercise combined with ChABC to promote functional recovery after spinal cord injury. We also present results of a preliminary study that tested the simplest approach for combining these treatments; use of a single intraparenchymal injection of ChABC administered to the lumbar enlargement of mice with voluntary wheel running exercise after a mid-thoracic spinal contusion injury. The results are negative, yet serve to highlight limitations in our understanding of the most effective protocols for combining these approaches. Further work is directed to identify the timing, type, and quantity of exercise and pharmacological interventions that can be used to maximize functional improvements by strengthening appropriate synaptic connections.
Collapse
Affiliation(s)
- Lyn B Jakeman
- Department of Physiology and Cell Biology, The Ohio State University Medical Center, Columbus, 43210, USA.
| | | | | |
Collapse
|
45
|
Gibbs KM, Chittur SV, Szaro BG. Metamorphosis and the regenerative capacity of spinal cord axons in Xenopus laevis. Eur J Neurosci 2010; 33:9-25. [PMID: 21059114 DOI: 10.1111/j.1460-9568.2010.07477.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Throughout the vertebrate subphylum, the regenerative potential of central nervous system axons is greatest in embryonic stages and declines as development progresses. For example, Xenopus laevis can functionally recover from complete transection of the spinal cord as a tadpole but is unable to do so after metamorphosing into a frog. Neurons of the reticular formation and raphe nucleus are among those that regenerate axons most reliably in tadpole and that lose this ability after metamorphosis. To identify molecular factors associated with the success and failure of spinal cord axon regeneration, we pharmacologically manipulated thyroid hormone (TH) levels using methimazole or triiodothyronine, to either keep tadpoles in a permanently larval state or induce precocious metamorphosis, respectively. Following complete spinal cord transection, serotonergic axons crossed the lesion site and tadpole swimming ability was restored when metamorphosis was inhibited, but these events failed to occur when metamorphosis was prematurely induced. Thus, the metamorphic events controlled by TH led directly to the loss of regenerative potential. Microarray analysis identified changes in hindbrain gene expression that accompanied regeneration-permissive and -inhibitory conditions, including many genes in the permissive condition that have been previously associated with axon outgrowth and neuroprotection. These data demonstrate that changes in gene expression occur within regenerating neurons in response to axotomy under regeneration-permissive conditions in which normal development has been suspended, and they identify candidate genes for future studies of how central nervous system axons can successfully regenerate in some vertebrates.
Collapse
Affiliation(s)
- Kurt M Gibbs
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY 12222, USA
| | | | | |
Collapse
|
46
|
Chang YW, Goff LA, Li H, Kane-Goldsmith N, Tzatzalos E, Hart RP, Young W, Grumet M. Rapid induction of genes associated with tissue protection and neural development in contused adult spinal cord after radial glial cell transplantation. J Neurotrauma 2010; 26:979-93. [PMID: 19257808 DOI: 10.1089/neu.2008.0762] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cell-based therapy has been widely evaluated in spinal cord injury (SCI) animal models and shown to improve functional recovery. However, host response to cell transplants at gene expression level is rarely discussed. We reported previously that acute transplantation of radial glial cells RG3.6 following SCI promoted early locomotion improvement within 1 week post-injury. To identify rapid molecular changes induced by RG3.6 transplantation in the host tissue, distal spinal cord segments were subjected to microarray analysis. Although RG3.6 transplantation, reduced activity of macrophages as early as 1-2 weeks post-injury, the expression levels of inflammatory genes (e.g., IL-6, MIP-2, MCP-1) were not decreased by RG3.6 treatment as compared to medium or other cell controls at 6-12 h post-injury. However, genes associated with tissue protection (Hsp70 and Hsp32) and neural cell development (Foxg1, Top2a, Sox11, Nkx2.2, Vimentin) were found to be significantly up-regulated by RG3.6 transplants. Foxg1 was the most highly induced gene in the RG3.6-treated spinal cords, and its expression by immunocytochemistry was confirmed in the host tissue. Moreover, RG3.6 treatment boosted the number of Nkx2.2 cells in the spinal cord, and these cells frequently co-expressed NG2, which marks progenitor cells. Taken together, these results demonstrate that radial glial transplants induced rapid and specific gene expression in the injured host tissue, and suggest that these early responses are associated with mechanisms of tissue protection and activation of endogenous neural progenitor cells.
Collapse
Affiliation(s)
- Yu-Wen Chang
- W.M. Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey 08854-8082, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Babiarz J, Kane-Goldsmith N, Basak S, Liu K, Young W, Grumet M. Juvenile and adult olfactory ensheathing cells bundle and myelinate dorsal root ganglion axons in culture. Exp Neurol 2010; 229:72-9. [PMID: 20850435 DOI: 10.1016/j.expneurol.2010.08.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 07/29/2010] [Accepted: 08/25/2010] [Indexed: 01/09/2023]
Abstract
Olfactory ensheathing cells (OEC), which normally associate closely with but do not myelinate axons in situ, myelinate axons in the adult mammalian spinal cord. They are of clinical interest as candidate cells for autologous transplantation but the ability of OEC to myelinate axons in vitro has been controversial. To clarify this issue, we isolated OEC from olfactory bulbs (OB) of juvenile and adult rats expressing GFP and analyzed their ability to myelinate axons. Using a well-defined assay for myelination of dorsal root ganglia (DRG) axons in culture, we found that OEC from juvenile pups associated with and then myelinated DRG axons. OEC assembled into bundles with the axons by 1week and required more than a week before myelination on axons was detected. In contrast, rat Schwann cells did not bundle axons and they formed P0(+) and MBP(+) myelin segments after as little as 1week. Most of the OEC in culture exhibited staining for calponin, a marker that was not found on Schwann cells in culture, whereas in both OEC and Schwann cell populations nearly all cells were positive for p75NTR and GFAP. These results confirm previous reports showing only subtle immunological differences between Schwann cells and OEC. Besides differences in the rate of myelination, we detected two additional functional differences in the interactions of OEC and Schwann cells with DRG axons. First, the diameter of OEC generated myelin was greater than for Schwann cell myelin on DRG axons. Second, OEC but not Schwann cells myelinated DRG axons in the absence of vitamin C. OEC isolated from adult OB were also found to bundle and myelinate DRG axons but the latter occurred only after incubation times of at least 3weeks. The results indicate that adult OEC require longer incubation times than juvenile OEC to myelinate axons and suggest that patterns of myelination by OEC and Schwann cells are distinguishable at least on axons in vitro. This article is part of a Special Issue entitled: Understanding olfactory ensheathing glia and their prospect for nervous system repair.
Collapse
Affiliation(s)
- Joanne Babiarz
- W.M. Keck Center for Collaborative Neuroscience, Rutgers, State University of New Jersey, 604 Allison Rd., Piscataway, NJ 08854-8082, USA
| | | | | | | | | | | |
Collapse
|
48
|
Endogenous radial glial cells support regenerating axons after spinal cord transection. Neuroreport 2010; 21:871-6. [DOI: 10.1097/wnr.0b013e32833d9695] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
49
|
Knerlich-Lukoschus F, von der Ropp-Brenner B, Lucius R, Mehdorn HM, Held-Feindt J. Chemokine expression in the white matter spinal cord precursor niche after force-defined spinal cord contusion injuries in adult rats. Glia 2010; 58:916-31. [PMID: 20155816 DOI: 10.1002/glia.20974] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Inflammatory cascades induced by spinal cord injuries (SCI) are localized in the white matter, a recognized neural stem- and progenitor-cell (NSPC) niche of the adult spinal cord. Chemokines, as integrators of these processes, might also be important determinants of this NSPC niche. CCL3/CCR1, CCL2/CCR2, and SDF-1alpha/CXCR4 were analyzed in the ventrolateral white matter after force defined thoracic SCI: Immunoreactivity (IR) density levels were measured 2 d, 7 d, 14 d, and 42 d on cervical (C 5), thoracic (T 5), and lumbar (L 5) levels. On day post operation (DPO) 42, chemokine inductions were further evaluated by real-time RT-PCR and Western blot analyses. Cellular phenotypes were confirmed by double labeling with markers for major cell types and NSPCs (nestin, Musashi-1, NG2, 3CB2, BLBP). Mitotic profiles were investigated in parallel by BrdU labeling. After lesion, chemokines were induced in the ventrolateral white matter on IR-, mRNA-, and protein-level. IR was generally more pronounced after severe lesions, with soaring increases of CCL2/CCR2 and continuous elevations of CCL3/CCR1. SDF-1alpha and CXCR4 IR induction was focused on thoracic levels. Chemokines/-receptors were co-expressed with astroglial, oligodendroglial markers, nestin, 3CB2 and BLBP by cells morphologically resembling radial glia on DPO 7 to DPO 42, and NG2 or Musashi-1 on DPO 2 and 7. In the white matter BrdU positive cells were significantly elevated after lesion compared with sham controls on all investigated time points peaking in the early time course on thoracic level: Here, chemokines were co-expressed by subsets of BrdU-labeled cells. These findings suggest an important role of chemokines/-receptors in the subpial white matter NSPC niche after SCI.
Collapse
|
50
|
Xu L, Xu CJ, Lü HZ, Wang YX, Li Y, Lu PH. Long-term fate of allogeneic neural stem cells following transplantation into injured spinal cord. Stem Cell Rev Rep 2010; 6:121-36. [PMID: 20012713 DOI: 10.1007/s12015-009-9104-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
To characterize the fate of allogeneic neural stem cells (NSCs) following transplantation into injured spinal cord, green fluorescent protein (GFP)-NSCs isolated from GFP transgenic Sprague-Dawley rat embryos were transplanted into contused spinal cords of Wistar rats. The GFP-NSCs survived for at least 6 months in injured spinal cord; most of them differentiated rapidly into astrocytes, and a few were able to undergo proliferation. After transplantation, the GFP-NSCs remained in the transplantation site at the early stage, and then migrated along white-matter, and gathered around the injured cavity. At 6 months post-transplantation, CD8 T-lymphocytes infiltrated the spinal cord, and mixed lymphocyte culture from host and donor showed that lymphocytes from the host spleen were primed by allogeneic GFP-NSCs. At 12 months post-transplantation, most GFP cells in the spinal cord lost their morphology and disintegrated. The Basso, Beattie and Bresnahan score and footprint analysis indicated that the improvement of locomotor function in transplanted rats appeared only at the early stage, and was not seen even at 6 months after transplantation All these results suggest that the allogeneic NSCs, after transplantation into injured spinal cord, activate the host immune system. Therefore, if immunosuppressive agents are not used, the grafted allogeneic NSCs, although they can survive for a long time, are subjected to host immune rejection, and the effect of NSCs on functional recovery is limited.
Collapse
Affiliation(s)
- Liang Xu
- Department of Neurobiology, Shanghai Jiaotong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, People's Republic of China
| | | | | | | | | | | |
Collapse
|