1
|
Tucker A, Baltazar A, Eisdorfer JT, Thackray JK, Vo K, Thomas H, Tandon A, Moses J, Singletary B, Gillespie T, Smith A, Pauken A, Nadella S, Pitonak M, Letchuman S, Jang J, Totty M, Jalufka FL, Aceves M, Adler AF, Maren S, Blackmon H, McCreedy DA, Abraira V, Dulin JN. Functional synaptic connectivity of engrafted spinal cord neurons with locomotor circuitry in the injured spinal cord. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.05.644402. [PMID: 40236108 PMCID: PMC11996546 DOI: 10.1101/2025.04.05.644402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Spinal cord injury (SCI) results in significant neurological deficits, with no currently available curative therapies. Neural progenitor cell (NPC) transplantation has emerged as a promising approach for neural repair, as graft-derived neurons (GDNs) can integrate into the host spinal cord and support axon regeneration. However, the mechanisms underlying functional recovery remain poorly understood. In this study, we investigate the synaptic integration of NPC-derived neurons into locomotor circuits, the projection patterns of distinct neuronal subtypes, and their potential to modulate motor circuit activity. Using transsynaptic tracing in a mouse thoracic contusion SCI model, we found that NPC-derived neurons form synaptic connections with host locomotor circuits, albeit at low frequencies. Furthermore, we mapped the axon projections of V0C and V2a interneurons, revealing distinct termination patterns within host spinal cord laminae. To assess functional integration, we employed chemogenetic activation of GDNs, which induced muscle activity in a subset of transplanted animals. However, NPC transplantation alone did not significantly improve locomotor recovery, highlighting a key challenge in the field. Our findings suggest that while GDNs can integrate into host circuits and modulate motor activity, synaptic connectivity remains a limiting factor in functional recovery. Future studies should focus on enhancing graft-host connectivity and optimizing transplantation strategies to maximize therapeutic benefits for SCI.
Collapse
|
2
|
Aceves M, Tucker A, Chen J, Vo K, Moses J, Amar Kumar P, Thomas H, Miranda D, Dampf G, Dietz V, Chang M, Lukose A, Jang J, Nadella S, Gillespie T, Trevino C, Buxton A, Pritchard AL, Green P, McCreedy DA, Dulin JN. Developmental stage of transplanted neural progenitor cells influences anatomical and functional outcomes after spinal cord injury in mice. Commun Biol 2023; 6:544. [PMID: 37208439 PMCID: PMC10199026 DOI: 10.1038/s42003-023-04893-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/02/2023] [Indexed: 05/21/2023] Open
Abstract
Neural progenitor cell (NPC) transplantation is a promising therapeutic strategy for replacing lost neurons following spinal cord injury (SCI). However, how graft cellular composition influences regeneration and synaptogenesis of host axon populations, or recovery of motor and sensory functions after SCI, is poorly understood. We transplanted developmentally-restricted spinal cord NPCs, isolated from E11.5-E13.5 mouse embryos, into sites of adult mouse SCI and analyzed graft axon outgrowth, cellular composition, host axon regeneration, and behavior. Earlier-stage grafts exhibited greater axon outgrowth, enrichment for ventral spinal cord interneurons and Group-Z spinal interneurons, and enhanced host 5-HT+ axon regeneration. Later-stage grafts were enriched for late-born dorsal horn interneuronal subtypes and Group-N spinal interneurons, supported more extensive host CGRP+ axon ingrowth, and exacerbated thermal hypersensitivity. Locomotor function was not affected by any type of NPC graft. These findings showcase the role of spinal cord graft cellular composition in determining anatomical and functional outcomes following SCI.
Collapse
Affiliation(s)
- Miriam Aceves
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA
| | - Ashley Tucker
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA
| | - Joseph Chen
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Katie Vo
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Joshua Moses
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | | | - Hannah Thomas
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Diego Miranda
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Gabrielle Dampf
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Valerie Dietz
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Matthew Chang
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Aleena Lukose
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Julius Jang
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Sneha Nadella
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Tucker Gillespie
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Christian Trevino
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
| | - Andrew Buxton
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Anna L Pritchard
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | | | - Dylan A McCreedy
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Jennifer N Dulin
- Department of Biology, Texas A&M University, College Station, TX, 77843, USA.
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
3
|
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
|
4
|
Pitonak M, Aceves M, Kumar PA, Dampf G, Green P, Tucker A, Dietz V, Miranda D, Letchuman S, Jonika MM, Bautista D, Blackmon H, Dulin JN. Effects of biological sex mismatch on neural progenitor cell transplantation for spinal cord injury in mice. Nat Commun 2022; 13:5380. [PMID: 36104357 PMCID: PMC9474813 DOI: 10.1038/s41467-022-33134-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 09/02/2022] [Indexed: 12/03/2022] Open
Abstract
Despite advancement of neural progenitor cell transplantation to spinal cord injury clinical trials, there remains a lack of understanding of how biological sex of transplanted cells influences outcomes after transplantation. To address this, we transplanted GFP-expressing sex-matched, sex-mismatched, or mixed donor cells into sites of spinal cord injury in adult male and female mice. Biological sex of the donor cells does not influence graft neuron density, glial differentiation, formation of the reactive glial cell border, or graft axon outgrowth. However, male grafts in female hosts feature extensive hypervascularization accompanied by increased vascular diameter and perivascular cell density. We show greater T-cell infiltration within male-to-female grafts than other graft types. Together, these findings indicate a biological sex-specific immune response of female mice to male donor cells. Our work suggests that biological sex should be considered in the design of future clinical trials for cell transplantation in human injury. In this study, Pitonak et al. report that transplantation of neural progenitor cells derived from male donors trigger an immune rejection response following transplantation into sites of spinal cord injury in female mice.
Collapse
|
5
|
Kiaie N, Gorabi AM, Loveless R, Teng Y, Jamialahmadi T, Sahebkar A. The regenerative potential of glial progenitor cells and reactive astrocytes in CNS injuries. Neurosci Biobehav Rev 2022; 140:104794. [PMID: 35902044 DOI: 10.1016/j.neubiorev.2022.104794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 10/16/2022]
Abstract
Cell therapeutic approaches focusing on the regeneration of damaged tissue have been a popular topic among researchers in recent years. In particular, self-repair scarring from the central nervous system (CNS) can significantly complicate the treatment of an injured patient. In CNS regeneration schemes, either glial progenitor cells or reactive glial cells have key roles to play. In this review, the contribution and underlying mechanisms of these progenitor/reactive glial cells during CNS regeneration are discussed, as well as their role in CNS-related diseases.
Collapse
Affiliation(s)
- Nasim Kiaie
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Armita Mahdavi Gorabi
- Department of Tissue Engineering and Applied Cell Science, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reid Loveless
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Yong Teng
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Tannaz Jamialahmadi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Baldassarri M, Buda R, Perazzo L, Ghinelli D, Sarino R, Grigolo B, Faldini C. Osteocondritis dissecans lesions of the knee restored by bone marrow aspirate concentrate. Clinical and imaging results in 18 patients. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY AND TRAUMATOLOGY 2022; 33:857-867. [PMID: 35133501 PMCID: PMC10126062 DOI: 10.1007/s00590-022-03214-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 01/24/2022] [Indexed: 10/19/2022]
Abstract
Abstract
Background
Osteochondritis dissecans (OCD) is a common cartilage disorder that specifically affects the knees of skeletally immature and young adult patients. There have been a few treatments that have been proposed: fixation of the fragment, drilling, microfractures. The aim of this study was to analyze retrospectively clinical and imaging results obtained by treating it with one-step bone marrow-derived cells Transplantation (BMDCT) technique.
Methods
From 2007 to 2014, 18 patients (mean-age 19.1 ± 5.0 years) affected by OCD were treated with one-step BMDC transplantation. In our observational study, clinical evaluation was performed at a scheduled follow-up through IKDC, Tegner, KOOS and EQ-VAS. X-rays and MRI were conducted preoperatively and at 12 months. At final follow-up, MRI MOCART Score was evaluated.
Results
IKDC and KOOS clinical scores showed a progressive increase. Tegner Score at final follow-up (5.3 ± 2.7) was significantly lower compared to the pre-injury level (6.5 ± 2.1); however, these results showed a statistically significant improvement that remained over time. EQ-VAS showed a significant improvement in every follow-up measure. MRI Mocart Score showed a complete or almost complete filling of the lesion in 13 patients.
Conclusions
“One-step” technique allows articular surface restoration with viable physiologic osteochondral tissue with a high clinical efficacy and imaging results. The number of cases is still limited, and further studies with larger sample sizes and greater follow-up evaluations are required to confirm our results. Nevertheless, we believe that BMDCT may represent a suitable option to treat OCD lesion in young adults.
Collapse
|
7
|
Long-Term Effects of Neural Precursor Cell Transplantation on Secondary Injury Processes and Functional Recovery after Severe Cervical Contusion-Compression Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms222313106. [PMID: 34884911 PMCID: PMC8658203 DOI: 10.3390/ijms222313106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/29/2021] [Accepted: 12/02/2021] [Indexed: 01/21/2023] Open
Abstract
Cervical spinal cord injury (SCI) remains a devastating event without adequate treatment options despite decades of research. In this context, the usefulness of common preclinical SCI models has been criticized. We, therefore, aimed to use a clinically relevant animal model of severe cervical SCI to assess the long-term effects of neural precursor cell (NPC) transplantation on secondary injury processes and functional recovery. To this end, we performed a clip contusion-compression injury at the C6 level in 40 female Wistar rats and a sham surgery in 10 female Wistar rats. NPCs, isolated from the subventricular zone of green fluorescent protein (GFP) expressing transgenic rat embryos, were transplanted ten days after the injury. Functional recovery was assessed weekly, and FluoroGold (FG) retrograde fiber-labeling, as well as manganese-enhanced magnetic resonance imaging (MEMRI), were performed prior to the sacrifice of the animals eight weeks after SCI. After cryosectioning of the spinal cords, immunofluorescence staining was conducted. Results were compared between the treatment groups (NPC, Vehicle, Sham) and statistically analyzed (p < 0.05 was considered significant). Despite the severity of the injury, leading to substantial morbidity and mortality during the experiment, long-term survival of the engrafted NPCs with a predominant differentiation into oligodendrocytes could be observed after eight weeks. While myelination of the injured spinal cord was not significantly improved, NPC treated animals showed a significant increase of intact perilesional motor neurons and preserved spinal tracts compared to untreated Vehicle animals. These findings were associated with enhanced preservation of intact spinal cord tissue. However, reactive astrogliosis and inflammation where not significantly reduced by the NPC-treatment. While differences in the Basso–Beattie–Bresnahan (BBB) score and the Gridwalk test remained insignificant, animals in the NPC group performed significantly better in the more objective CatWalk XT gait analysis, suggesting some beneficial effects of the engrafted NPCs on the functional recovery after severe cervical SCI.
Collapse
|
8
|
Zholudeva LV, Jin Y, Qiang L, Lane MA, Fischer I. Preparation of Neural Stem Cells and Progenitors: Neuronal Production and Grafting Applications. Methods Mol Biol 2021; 2311:73-108. [PMID: 34033079 PMCID: PMC10074836 DOI: 10.1007/978-1-0716-1437-2_7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Neural stem cells (NSCs) are a valuable tool for the study of neural development and function as well as an important source of cell transplantation strategies for neural disease. NSCs can be used to study how neurons acquire distinct phenotypes and how the interactions between neurons and glial cells in the developing nervous system shape the structure and function of the CNS. NSCs can also be used for cell replacement therapies following CNS injury targeting astrocytes, oligodendrocytes, and neurons. With the availability of patient-derived induced pluripotent stem cells (iPSCs), neurons prepared from NSCs can be used to elucidate the molecular basis of neurological disorders leading to potential treatments. Although NSCs can be derived from different species and many sources, including embryonic stem cells (ESCs), iPSCs, adult CNS, and direct reprogramming of nonneural cells, isolating primary NSCs directly from fetal tissue is still the most common technique for preparation and study of neurons. Regardless of the source of tissue, similar techniques are used to maintain NSCs in culture and to differentiate NSCs toward mature neural lineages. This chapter will describe specific methods for isolating and characterizing multipotent NSCs and neural precursor cells (NPCs) from embryonic rat CNS tissue (mostly spinal cord) and from human ESCs and iPSCs as well as NPCs prepared by reprogramming. NPCs can be separated into neuronal and glial restricted progenitors (NRP and GRP, respectively) and used to reliably produce neurons or glial cells both in vitro and following transplantation into the adult CNS. This chapter will describe in detail the methods required for the isolation, propagation, storage, and differentiation of NSCs and NPCs isolated from rat and mouse spinal cords for subsequent in vitro or in vivo studies as well as new methods associated with ESCs, iPSCs, and reprogramming.
Collapse
Affiliation(s)
- Lyandysha V Zholudeva
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Ying Jin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Liang Qiang
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Bonilla C, Zurita M. Cell-Based Therapies for Traumatic Brain Injury: Therapeutic Treatments and Clinical Trials. Biomedicines 2021; 9:biomedicines9060669. [PMID: 34200905 PMCID: PMC8230536 DOI: 10.3390/biomedicines9060669] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic brain injury (TBI) represents physical damage to the brain tissue that induces transitory or permanent neurological disabilities. TBI contributes to 50% of all trauma deaths, with many enduring long-term consequences and significant medical and rehabilitation costs. There is currently no therapy to reverse the effects associated with TBI. An increasing amount of research has been undertaken regarding the use of different stem cells (SCs) to treat the consequences of brain damage. Neural stem cells (NSCs) (adult and embryonic) and mesenchymal stromal cells (MSCs) have shown efficacy in pre-clinical models of TBI and in their introduction to clinical research. The purpose of this review is to provide an overview of TBI and the state of clinical trials aimed at evaluating the use of stem cell-based therapies in TBI. The primary aim of these studies is to investigate the safety and efficacy of the use of SCs to treat this disease. Although an increasing number of studies are being carried out, few results are currently available. In addition, we present our research regarding the use of cell therapy in TBI. There is still a significant lack of understanding regarding the cell therapy mechanisms for the treatment of TBI. Thus, future studies are needed to evaluate the feasibility of the transplantation of SCs in TBI.
Collapse
Affiliation(s)
- Celia Bonilla
- Cell Therapy Unit, Puerta de Hierro Hospital, 28222 Majadahonda, Madrid, Spain
- Correspondence: ; Tel.: +34-91-191-7879
| | - Mercedes Zurita
- Cell Therapy Unit Responsable, Puerta de Hierro Hospital, 28222 Majadahonda, Madrid, Spain;
| |
Collapse
|
10
|
Fischer I, Dulin JN, Lane MA. Transplanting neural progenitor cells to restore connectivity after spinal cord injury. Nat Rev Neurosci 2020; 21:366-383. [PMID: 32518349 PMCID: PMC8384139 DOI: 10.1038/s41583-020-0314-2] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2020] [Indexed: 12/12/2022]
Abstract
Spinal cord injury remains a scientific and therapeutic challenge with great cost to individuals and society. The goal of research in this field is to find a means of restoring lost function. Recently we have seen considerable progress in understanding the injury process and the capacity of CNS neurons to regenerate, as well as innovations in stem cell biology. This presents an opportunity to develop effective transplantation strategies to provide new neural cells to promote the formation of new neuronal networks and functional connectivity. Past and ongoing clinical studies have demonstrated the safety of cell therapy, and preclinical research has used models of spinal cord injury to better elucidate the underlying mechanisms through which donor cells interact with the host and thus increase long-term efficacy. While a variety of cell therapies have been explored, we focus here on the use of neural progenitor cells obtained or derived from different sources to promote connectivity in sensory, motor and autonomic systems.
Collapse
Affiliation(s)
- Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Jennifer N Dulin
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
11
|
Willing AE, Das M, Howell M, Mohapatra SS, Mohapatra S. Potential of mesenchymal stem cells alone, or in combination, to treat traumatic brain injury. CNS Neurosci Ther 2020; 26:616-627. [PMID: 32157822 PMCID: PMC7248546 DOI: 10.1111/cns.13300] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/17/2020] [Accepted: 02/23/2020] [Indexed: 12/13/2022] Open
Abstract
Traumatic brain injury (TBI) causes death and disability in the United States and around the world. The traumatic insult causes the mechanical injury of the brain and primary cellular death. While a comprehensive pathological mechanism of TBI is still lacking, the focus of the TBI research is concentrated on understanding the pathophysiology and developing suitable therapeutic approaches. Given the complexities in pathophysiology involving interconnected immunologic, inflammatory, and neurological cascades occurring after TBI, the therapies directed to a single mechanism fail in the clinical trials. This has led to the development of the paradigm of a combination therapeutic approach against TBI. While there are no drugs available for the treatment of TBI, stem cell therapy has shown promising results in preclinical studies. But, the success of the therapy depends on the survival of the stem cells, which are limited by several factors including route of administration, health of the administered cells, and inflammatory microenvironment of the injured brain. Reducing the inflammation prior to cell administration may provide a better outcome of cell therapy following TBI. This review is focused on different therapeutic approaches of TBI and the present status of the clinical trials.
Collapse
Affiliation(s)
- Alison E Willing
- Department of Neurosurgery and Brain Repair, Center of Excellence for Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Mahasweta Das
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA.,James A. Haley Veterans Hospital, Tampa, FL, USA
| | - Mark Howell
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA.,James A. Haley Veterans Hospital, Tampa, FL, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Hospital, Tampa, FL, USA.,Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Subhra Mohapatra
- Department of Molecular Medicine, University of South Florida Morsani College of Medicine, Tampa, FL, USA.,James A. Haley Veterans Hospital, Tampa, FL, USA
| |
Collapse
|
12
|
Adler AF, Björklund A, Parmar M. Transsynaptic tracing and its emerging use to assess graft-reconstructed neural circuits. Stem Cells 2020; 38:716-726. [PMID: 32101353 DOI: 10.1002/stem.3166] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/20/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022]
Abstract
Fetal neural progenitor grafts have been evaluated in preclinical animal models of spinal cord injury and Parkinson's disease for decades, but the initial reliance on primary tissue as a cell source limited the scale of their clinical translatability. With the development of robust methods to differentiate human pluripotent stem cells to specific neural subtypes, cell replacement therapy holds renewed promise to treat a variety of neurodegenerative diseases and injuries at scale. As these cell sources are evaluated in preclinical models, new transsynaptic tracing methods are making it possible to study the connectivity between host and graft neurons with greater speed and detail than was previously possible. To date, these studies have revealed that widespread, long-lasting, and anatomically appropriate synaptic contacts are established between host and graft neurons, as well as new aspects of host-graft connectivity which may be relevant to clinical cell replacement therapy. It is not yet clear, however, whether the synaptic connectivity between graft and host neurons is as cell-type specific as it is in the endogenous nervous system, or whether that connectivity is responsible for the functional efficacy of cell replacement therapy. Here, we review evidence suggesting that the new contacts established between host and graft neurons may indeed be cell-type specific, and how transsynaptic tracing can be used in the future to further elucidate the mechanisms of graft-mediated functional recovery in spinal cord injury and Parkinson's disease.
Collapse
Affiliation(s)
- Andrew F Adler
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Anders Björklund
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Malin Parmar
- Developmental and Regenerative Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.,Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
13
|
Safe nanoengineering and incorporation of transplant populations in a neurosurgical grade biomaterial, DuraGen Plus TM, for protected cell therapy applications. J Control Release 2020; 321:553-563. [PMID: 32087299 DOI: 10.1016/j.jconrel.2020.02.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/05/2020] [Accepted: 02/17/2020] [Indexed: 11/22/2022]
Abstract
High transplant cell loss is a major barrier to translation of stem cell therapy for pathologies of the brain and spinal cord. Encapsulated delivery of stem cells in biomaterials for cell therapy is gaining popularity but experimental research has overwhelmingly used laboratory grade materials unsuitable for human clinical use - representing a further barrier to clinical translation. A potential solution is to use neurosurgical grade materials routinely used in clinical protocols which have an established human safety profile. Here, we tested the ability of Duragen Plus™ - a clinical biomaterial used widely in neurosurgical duraplasty procedures, to support the growth and differentiation of neural stem cells- a major transplant population being tested in clinical trials for neurological pathology. Genetic engineering of stem cells yields augmented therapeutic cells, so we further tested the ability of the Duragen Plus™ matrix to support stem cells engineered using magnetofection technology and minicircle DNA vectors- a promising cell engineering approach we previously reported (Journal of Controlled Release, 2016 a &b). The safety of the nano-engineering approach was analysed for the first time using sophisticated data-independent analysis by mass spectrometry-based proteomics. We prove that the Duragen Plus™ matrix is a promising biomaterial for delivery of stem cell transplant populations, with no adverse effects on key regenerative parameters. This advanced cellular construct based on a combinatorial nano-engineering and biomaterial encapsulation approach, could therefore offer key advantages for clinical translation.
Collapse
|
14
|
Astrocytes migrate from human neural stem cell grafts and functionally integrate into the injured rat spinal cord. Exp Neurol 2019; 314:46-57. [DOI: 10.1016/j.expneurol.2019.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/13/2018] [Accepted: 01/12/2019] [Indexed: 11/21/2022]
|
15
|
Payne SL, Tuladhar A, Obermeyer JM, Varga BV, Teal CJ, Morshead CM, Nagy A, Shoichet MS. Initial cell maturity changes following transplantation in a hyaluronan-based hydrogel and impacts therapeutic success in the stroke-injured rodent brain. Biomaterials 2019; 192:309-322. [DOI: 10.1016/j.biomaterials.2018.11.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/31/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022]
|
16
|
Spruance VM, Zholudeva LV, Hormigo KM, Randelman ML, Bezdudnaya T, Marchenko V, Lane MA. Integration of Transplanted Neural Precursors with the Injured Cervical Spinal Cord. J Neurotrauma 2018; 35:1781-1799. [PMID: 29295654 PMCID: PMC6033309 DOI: 10.1089/neu.2017.5451] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cervical spinal cord injuries (SCI) result in devastating functional consequences, including respiratory dysfunction. This is largely attributed to the disruption of phrenic pathways, which control the diaphragm. Recent work has identified spinal interneurons as possible contributors to respiratory neuroplasticity. The present work investigated whether transplantation of developing spinal cord tissue, inherently rich in interneuronal progenitors, could provide a population of new neurons and growth-permissive substrate to facilitate plasticity and formation of novel relay circuits to restore input to the partially denervated phrenic motor circuit. One week after a lateralized, C3/4 contusion injury, adult Sprague-Dawley rats received allografts of dissociated, developing spinal cord tissue (from rats at gestational days 13-14). Neuroanatomical tracing and terminal electrophysiology was performed on the graft recipients 1 month later. Experiments using pseudorabies virus (a retrograde, transynaptic tracer) revealed connections from donor neurons onto host phrenic circuitry and from host, cervical interneurons onto donor neurons. Anatomical characterization of donor neurons revealed phenotypic heterogeneity, though donor-host connectivity appeared selective. Despite the consistent presence of cholinergic interneurons within donor tissue, transneuronal tracing revealed minimal connectivity with host phrenic circuitry. Phrenic nerve recordings revealed changes in burst amplitude after application of a glutamatergic, but not serotonergic antagonist to the transplant, suggesting a degree of functional connectivity between donor neurons and host phrenic circuitry that is regulated by glutamatergic input. Importantly, however, anatomical and functional results were variable across animals, and future studies will explore ways to refine donor cell populations and entrain consistent connectivity.
Collapse
Affiliation(s)
- Victoria M Spruance
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Lyandysha V Zholudeva
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Kristiina M Hormigo
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Margo L Randelman
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Tatiana Bezdudnaya
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Vitaliy Marchenko
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine , Philadelphia, Pennsylvania
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Spinal Cord Research Center, Drexel University College of Medicine , Philadelphia, Pennsylvania
| |
Collapse
|
17
|
Wu D, Zhang Y, Xu X, Guo T, Xie D, Zhu R, Chen S, Ramakrishna S, He L. RGD/TAT-functionalized chitosan-graft-PEI-PEG gene nanovector for sustained delivery of NT-3 for potential application in neural regeneration. Acta Biomater 2018; 72:266-277. [PMID: 29578088 DOI: 10.1016/j.actbio.2018.03.030] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 03/11/2018] [Accepted: 03/15/2018] [Indexed: 11/18/2022]
Abstract
In this study, we prepared a multifunctional gene delivery nanovector containing a chitosan (CS) backbone and polyethylenimine (PEI) arms with arginine-glycine-aspartate (RGD)/twin-arginine translocation (TAT) conjugated via polyethylene glycol (PEG). Branched PEI, with a molecular weight of 2000 Da, was used to achieve a balance between biocompatibility and transfection efficiency, whereas RGD/TAT peptides were conjugated for enhanced targeting ability and cellular uptake. Synthesis of the copolymers was confirmed by characterizing the chemical structure with 1H nuclear magnetic resonance and Fourier Transform Infrared Spectroscopy (FTIR). The nanovector was biocompatible with cells and showed excellent capability for DNA condensation; the resulting complexes with DNA were well-formed, and possessed small particle size and reasonable positive charge. Higher gene transfection efficiency, compared to that achieved with PEI (25 kDa), was confirmed in tumor (HeLa cells) and normal cells (293T and NIH 3T3 cells). More importantly, the cells transfected with the chitosan-graft-PEI-PEG/pCMV-EGFP-Ntf3 complex produced sustained neurotrophin-3 with a linear increase in cumulative concentration, which induced neuronal differentiation of neural stem cell and promoted neurite outgrowth. These findings suggested that our multifunctional copolymers might be ideal nanovectors for engineering cells via gene transfection, and could potentially be applied in tumor therapy and regenerative medicine. STATEMENT OF SIGNIFICANCE We successfully prepared a multifunctional gene delivery nanovector containing branched PEI with a molecular weight of 2000 Da to balance between biocompatibility and transfection efficiency, and RGD/TAT peptides for enhanced targeting ability and cellular uptake. The well-formed CPPP/DNA complexes of small particle size and reasonable positive charges potentially enhanced gene transfection in both tumor and normal cells. More importantly, the CPPP/pCMV-EGFP-Ntf3 complex-transfected 293T cells could produce sustained NT-3 with a constant ratio, which induced neuron differentiation of NSC and promoted neurite outgrowth. Therefore, our study provided an effective strategy for producing neurotrophins by engineering cells with gene delivery, which deserved wide investigation and potential application in regenerative medicine.
Collapse
Affiliation(s)
- Dongni Wu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Yongnu Zhang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China
| | - Xiaoting Xu
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Ting Guo
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Deming Xie
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Rong Zhu
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Shengfeng Chen
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China
| | - Seeram Ramakrishna
- MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China; Guangdong-Hong Kong-Macau Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou 510632, China; Department of Mechanical Engineering, Faculty of Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Liumin He
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; MOE Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
18
|
Liu S, Schackel T, Weidner N, Puttagunta R. Biomaterial-Supported Cell Transplantation Treatments for Spinal Cord Injury: Challenges and Perspectives. Front Cell Neurosci 2018; 11:430. [PMID: 29375316 PMCID: PMC5768640 DOI: 10.3389/fncel.2017.00430] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/20/2017] [Indexed: 12/17/2022] Open
Abstract
Spinal cord injury (SCI), resulting in para- and tetraplegia caused by the partial or complete disruption of descending motor and ascending sensory neurons, represents a complex neurological condition that remains incurable. Following SCI, numerous obstacles comprising of the loss of neural tissue (neurons, astrocytes, and oligodendrocytes), formation of a cavity, inflammation, loss of neuronal circuitry and function must be overcome. Given the multifaceted primary and secondary injury events that occur with SCI treatment options are likely to require combinatorial therapies. While several methods have been explored, only the intersection of two, cell transplantation and biomaterial implantation, will be addressed in detail here. Owing to the constant advance of cell culture technologies, cell-based transplantation has come to the forefront of SCI treatment in order to replace/protect damaged tissue and provide physical as well as trophic support for axonal regrowth. Biomaterial scaffolds provide cells with a protected environment from the surrounding lesion, in addition to bridging extensive damage and providing physical and directional support for axonal regrowth. Moreover, in this combinatorial approach cell transplantation improves scaffold integration and therefore regenerative growth potential. Here, we review the advances in combinatorial therapies of Schwann cells (SCs), astrocytes, olfactory ensheathing cells (OECs), mesenchymal stem cells, as well as neural stem and progenitor cells (NSPCs) with various biomaterial scaffolds.
Collapse
Affiliation(s)
- Shengwen Liu
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Thomas Schackel
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
19
|
Payne SL, Anandakumaran PN, Varga BV, Morshead CM, Nagy A, Shoichet MS. In Vitro Maturation of Human iPSC-Derived Neuroepithelial Cells Influences Transplant Survival in the Stroke-Injured Rat Brain. Tissue Eng Part A 2017; 24:351-360. [PMID: 28594288 DOI: 10.1089/ten.tea.2016.0515] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Stem cell transplantation is a promising strategy for brain tissue regeneration; yet, despite some success, cell survival following transplantation remains low. In this study, we demonstrate that cell viability is enhanced by control over maturation of neuronal precursor cells, which are delivered in an injectable blend of hyaluronan and methylcellulose. We selected three subpopulations of human neuronal precursor cells derived from a cortically specified neuroepithelial stem cell (cNESC) population based on differences in expression of multipotent and neuron-specific proteins: early-, mid-, and late-differentiated neurons. These cells were transplanted into an endothelin-1 stroke-injured rat brain and their survival and fate were investigated 1 week later. Significantly, more cells were found in the brain after transplanting early- or mid- differentiated cNESCs compared to the late-differentiated population. The mid-differentiated population also had significantly more β-III tubulin-positive cells than either the early- or late-differentiated populations. These results suggest that maturity has a significant impact on cell survival following transplantation and cells with an intermediate maturity differentiate to neurons.
Collapse
Affiliation(s)
- Samantha L Payne
- 1 Department of Chemical Engineering and Applied Chemistry, University of Toronto , Toronto, Canada .,2 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Canada
| | - Priya N Anandakumaran
- 2 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Canada
| | - Balazs V Varga
- 3 Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital, Toronto, Canada
| | - Cindi M Morshead
- 4 Institute of Medical Science, University of Toronto , Toronto Canada
| | - Andras Nagy
- 3 Lunenfeld-Tanenbaum Research Institute , Mount Sinai Hospital, Toronto, Canada
| | - Molly S Shoichet
- 1 Department of Chemical Engineering and Applied Chemistry, University of Toronto , Toronto, Canada .,2 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, Canada .,5 Department of Chemistry, University of Toronto , Toronto, Canada
| |
Collapse
|
20
|
Charsar BA, Urban MW, Lepore AC. Harnessing the power of cell transplantation to target respiratory dysfunction following spinal cord injury. Exp Neurol 2016; 287:268-275. [PMID: 27531634 DOI: 10.1016/j.expneurol.2016.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 07/29/2016] [Accepted: 08/12/2016] [Indexed: 12/13/2022]
Abstract
The therapeutic benefit of cell transplantation has been assessed in a host of central nervous system (CNS) diseases, including disorders of the spinal cord such as traumatic spinal cord injury (SCI). The promise of cell transplantation to preserve and/or restore normal function can be aimed at a variety of therapeutic mechanisms, including replacement of lost or damaged CNS cell types, promotion of axonal regeneration or sprouting, neuroprotection, immune response modulation, and delivery of gene products such as neurotrophic factors, amongst other possibilities. Despite significant work in the field of transplantation in models of SCI, limited attention has been directed at harnessing the therapeutic potential of cell grafting for preserving respiratory function after SCI, despite the critical role pulmonary compromise plays in patient outcome in this devastating disease. Here, we will review the limited number of studies that have demonstrated the therapeutic potential of intraspinal transplantation of a variety of cell types for addressing respiratory dysfunction in SCI.
Collapse
Affiliation(s)
- Brittany A Charsar
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College, Thomas Jefferson University, 900 Walnut Street, JHN 418, Philadelphia, PA, 19107, United States
| | - Mark W Urban
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College, Thomas Jefferson University, 900 Walnut Street, JHN 418, Philadelphia, PA, 19107, United States
| | - Angelo C Lepore
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College, Thomas Jefferson University, 900 Walnut Street, JHN 418, Philadelphia, PA, 19107, United States.
| |
Collapse
|
21
|
Hayakawa K, Haas C, Fischer I. Examining the properties and therapeutic potential of glial restricted precursors in spinal cord injury. Neural Regen Res 2016; 11:529-33. [PMID: 27212899 PMCID: PMC4870895 DOI: 10.4103/1673-5374.180725] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In the aftermath of spinal cord injury, glial restricted precursors (GRPs) and immature astrocytes offer the potential to modulate the inflammatory environment of the injured spinal cord and promote host axon regeneration. Nevertheless clinical application of cellular therapy for the repair of spinal cord injury requires strict quality-assured protocols for large-scale production and preservation that necessitates long-term in vitro expansion. Importantly, such processes have the potential to alter the phenotypic and functional properties and thus therapeutic potential of these cells. Furthermore, clinical use of cellular therapies may be limited by the inflammatory microenvironment of the injured spinal cord, altering the phenotypic and functional properties of grafted cells. This report simulates the process of large-scale GRP production and demonstrates the permissive properties of GRP following long-term in vitro culture. Furthermore, we defined the phenotypic and functional properties of GRP in the presence of inflammatory factors, and call attention to the importance of the microenvironment of grafted cells, underscoring the importance of modulating the environment of the injured spinal cord.
Collapse
Affiliation(s)
- Kazuo Hayakawa
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Christopher Haas
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Itzhak Fischer
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| |
Collapse
|
22
|
Maclean FL, Rodriguez AL, Parish CL, Williams RJ, Nisbet DR. Integrating Biomaterials and Stem Cells for Neural Regeneration. Stem Cells Dev 2016; 25:214-26. [DOI: 10.1089/scd.2015.0314] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Francesca L. Maclean
- Research School of Engineering, the Australian National University, Canberra, Australia
| | | | - Clare L. Parish
- Florey Institute of Neuroscience and Mental Health, the University of Melbourne, Parkville, Australia
| | - Richard J. Williams
- School of Aerospace, Mechanical and Manufacturing Engineering and Health Innovations Research Institute, RMIT University, Melbourne, Australia
| | - David R. Nisbet
- Research School of Engineering, the Australian National University, Canberra, Australia
| |
Collapse
|
23
|
Li K, Javed E, Scura D, Hala TJ, Seetharam S, Falnikar A, Richard JP, Chorath A, Maragakis NJ, Wright MC, Lepore AC. Human iPS cell-derived astrocyte transplants preserve respiratory function after spinal cord injury. Exp Neurol 2015. [PMID: 26216662 DOI: 10.1016/j.expneurol.2015.07.020] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Transplantation-based replacement of lost and/or dysfunctional astrocytes is a promising therapy for spinal cord injury (SCI) that has not been extensively explored, despite the integral roles played by astrocytes in the central nervous system (CNS). Induced pluripotent stem (iPS) cells are a clinically-relevant source of pluripotent cells that both avoid ethical issues of embryonic stem cells and allow for homogeneous derivation of mature cell types in large quantities, potentially in an autologous fashion. Despite their promise, the iPS cell field is in its infancy with respect to evaluating in vivo graft integration and therapeutic efficacy in SCI models. Astrocytes express the major glutamate transporter, GLT1, which is responsible for the vast majority of glutamate uptake in spinal cord. Following SCI, compromised GLT1 expression/function can increase susceptibility to excitotoxicity. We therefore evaluated intraspinal transplantation of human iPS cell-derived astrocytes (hIPSAs) following cervical contusion SCI as a novel strategy for reconstituting GLT1 expression and for protecting diaphragmatic respiratory neural circuitry. Transplant-derived cells showed robust long-term survival post-injection and efficiently differentiated into astrocytes in injured spinal cord of both immunesuppressed mice and rats. However, the majority of transplant-derived astrocytes did not express high levels of GLT1, particularly at early times post-injection. To enhance their ability to modulate extracellular glutamate levels, we engineered hIPSAs with lentivirus to constitutively express GLT1. Overexpression significantly increased GLT1 protein and functional GLT1-mediated glutamate uptake levels in hIPSAs both in vitro and in vivo post-transplantation. Compared to human fibroblast control and unmodified hIPSA transplantation, GLT1-overexpressing hIPSAs reduced (1) lesion size within the injured cervical spinal cord, (2) morphological denervation by respiratory phrenic motor neurons at the diaphragm neuromuscular junction, and (3) functional diaphragm denervation as measured by recording of spontaneous EMGs and evoked compound muscle action potentials. Our findings demonstrate that hiPSA transplantation is a therapeutically-powerful approach for SCI.
Collapse
Affiliation(s)
- Ke Li
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College at Thomas Jefferson University, 900 Walnut Street, JHN 469, Philadelphia, PA 19107, United States.
| | - Elham Javed
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College at Thomas Jefferson University, 900 Walnut Street, JHN 469, Philadelphia, PA 19107, United States.
| | - Daniel Scura
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College at Thomas Jefferson University, 900 Walnut Street, JHN 469, Philadelphia, PA 19107, United States.
| | - Tamara J Hala
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College at Thomas Jefferson University, 900 Walnut Street, JHN 469, Philadelphia, PA 19107, United States.
| | - Suneil Seetharam
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College at Thomas Jefferson University, 900 Walnut Street, JHN 469, Philadelphia, PA 19107, United States.
| | - Aditi Falnikar
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College at Thomas Jefferson University, 900 Walnut Street, JHN 469, Philadelphia, PA 19107, United States.
| | - Jean-Philippe Richard
- Department of Neurology, Johns Hopkins University School of Medicine, 855N. Wolfe St., Rangos 250, Baltimore, MD 21205, United States.
| | - Ashley Chorath
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College at Thomas Jefferson University, 900 Walnut Street, JHN 469, Philadelphia, PA 19107, United States.
| | - Nicholas J Maragakis
- Department of Neurology, Johns Hopkins University School of Medicine, 855N. Wolfe St., Rangos 250, Baltimore, MD 21205, United States.
| | - Megan C Wright
- Department of Biology, Arcadia University, 450S. Easton Rd., 220 Boyer Hall, Glenside, PA 19038, United States.
| | - Angelo C Lepore
- Department of Neuroscience, Farber Institute for Neurosciences, Sidney Kimmel Medical College at Thomas Jefferson University, 900 Walnut Street, JHN 469, Philadelphia, PA 19107, United States.
| |
Collapse
|
24
|
Bonner JF, Steward O. Repair of spinal cord injury with neuronal relays: From fetal grafts to neural stem cells. Brain Res 2015; 1619:115-23. [PMID: 25591483 DOI: 10.1016/j.brainres.2015.01.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 12/29/2014] [Accepted: 01/05/2015] [Indexed: 02/01/2023]
Abstract
Spinal cord injury (SCI) disrupts the long axonal tracts of the spinal cord leading to devastating loss of function. Cell transplantation in the injured spinal cord has the potential to lead to recovery after SCI via a variety of mechanisms. One such strategy is the formation of neuronal relays between injured long tract axons and denervated neurons. The idea of creating a neuronal relay was first proposed over 25 years ago when fetal tissue was first successfully transplanted into the injured rodent spinal cord. Advances in labeling of grafted cells and the development of neural stem cell culturing techniques have improved the ability to create and refine such relays. Several recent studies have examined the ability to create a novel neuronal circuit between injured axons and denervated targets. This approach is an alternative to long-distance regeneration of damaged axons that may provide a meaningful degree of recovery without direct recreation of lost pathways. This brief review will examine the contribution of fetal grafting to current advances in neuronal grafting. Of particular interest will be the ability of transplanted neurons derived from fetal grafts, neural precursor cells and neural stem cells to reconnect long distance motor and sensory pathways of the injured spinal cord. This article is part of a Special Issue entitled SI: Spinal cord injury.
Collapse
Affiliation(s)
- Joseph F Bonner
- Reeve-Irvine Research Center, University of California, 1105 Gillespie Neuroscience Research Facility, Irvine, CA 92697-4265, USA.
| | - Oswald Steward
- Reeve-Irvine Research Center, University of California, 1105 Gillespie Neuroscience Research Facility, Irvine, CA 92697-4265, USA; Departments of Anatomy & Neurobiology, Neurobiology & Behavior, and Neurosurgery, University of California at Irvine School of Medicine, Irvine, CA 92697-4265, USA
| |
Collapse
|
25
|
Transplantation of glial progenitors that overexpress glutamate transporter GLT1 preserves diaphragm function following cervical SCI. Mol Ther 2014; 23:533-48. [PMID: 25492561 DOI: 10.1038/mt.2014.236] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 12/02/2014] [Indexed: 01/14/2023] Open
Abstract
Approximately half of traumatic spinal cord injury (SCI) cases affect cervical regions, resulting in chronic respiratory compromise. The majority of these injuries affect midcervical levels, the location of phrenic motor neurons (PMNs) that innervate the diaphragm. A valuable opportunity exists following SCI for preventing PMN loss that occurs during secondary degeneration. One of the primary causes of secondary injury is excitotoxicity due to dysregulation of extracellular glutamate homeostasis. Astrocytes express glutamate transporter 1 (GLT1), which is responsible for the majority of CNS glutamate clearance. Given our observations of GLT1 dysfunction post-SCI, we evaluated intraspinal transplantation of Glial-Restricted Precursors (GRPs)--a class of lineage-restricted astrocyte progenitors--into ventral horn following cervical hemicontusion as a novel strategy for reconstituting GLT1 function, preventing excitotoxicity and protecting PMNs in the acutely injured spinal cord. We find that unmodified transplants express low levels of GLT1 in the injured spinal cord. To enhance their therapeutic properties, we engineered GRPs with AAV8 to overexpress GLT1 only in astrocytes using the GFA2 promoter, resulting in significantly increased GLT1 protein expression and functional glutamate uptake following astrocyte differentiation in vitro and after transplantation into C4 hemicontusion. Compared to medium-only control and unmodified GRPs, GLT1-overexpressing transplants reduced lesion size, diaphragm denervation and diaphragm dysfunction. Our findings demonstrate transplantation-based replacement of astrocyte GLT1 is a promising approach for SCI.
Collapse
|
26
|
Konyalioglu S, Armagan G, Yalcin A, Atalayin C, Dagci T. Effects of resveratrol on hydrogen peroxide-induced oxidative stress in embryonic neural stem cells. Neural Regen Res 2014; 8:485-95. [PMID: 25206691 PMCID: PMC4146049 DOI: 10.3969/j.issn.1673-5374.2013.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 01/05/2013] [Indexed: 12/26/2022] Open
Abstract
Resveratrol, a natural phenolic compound, has been shown to prevent cardiovascular diseases and cancer and exhibit neuroprotective effects. In this study, we examined the neuroprotective and antioxidant effects of resveratrol against hydrogen peroxide in embryonic neural stem cells. Hydrogen peroxide treatment alone increased catalase and glutathione peroxidase activities but did not change superoxide dismutase levels compared with hydrogen peroxide + resveratrol treatment. Nitric oxide synthase activity and concomitant nitric oxide levels increased in response to hydrogen peroxide treatment. Conversely, resveratrol treatment decreased nitric oxide synthase activity and nitric oxide levels. Resveratrol also attenuated hydrogen peroxide-induced nuclear or mitochondrial DNA damage. We propose that resveratrol may be a promising agent for protecting embryonic neural stem cells because of its potential to decrease oxidative stress by inducing higher activity of antioxidant enzymes, decreasing nitric oxide production and nitric oxide synthase activity, and alleviating both nuclear and mitochondrial DNA damage.
Collapse
Affiliation(s)
- Sibel Konyalioglu
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Bornova-Izmir 35100, Turkey
| | - Guliz Armagan
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Bornova-Izmir 35100, Turkey
| | - Ayfer Yalcin
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Bornova-Izmir 35100, Turkey
| | - Cigdem Atalayin
- Department of Restorative Dentistry and Endodontics, Faculty of Dentistry, Ege University, Bornova-Izmir 35100, Turkey
| | - Taner Dagci
- Department of Physiology, School of Medicine, Ege University, Bornova-Izmir 35100, Turkey ; Center for Brain Research, Ege University, Bornova-Izmir 35100, Turkey
| |
Collapse
|
27
|
Shaltouki A, Peng J, Liu Q, Rao MS, Zeng X. Efficient generation of astrocytes from human pluripotent stem cells in defined conditions. Stem Cells 2014; 31:941-52. [PMID: 23341249 DOI: 10.1002/stem.1334] [Citation(s) in RCA: 194] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 12/24/2012] [Indexed: 01/07/2023]
Abstract
Astrocytes can be generated from various tissue sources including human pluripotent stem cells (PSC). In this manuscript, we describe a chemically defined xeno-free medium culture system for rapidly generating astrocytes from neural stem cells derived from PSC. We show that astrocyte development in vitro, mimics normal development in vivo, and also passes through a CD44(+) astrocyte precursor stage. Astrocytes generated by our method display similar gene expression patterns, morphological characteristics and functional properties to primary astrocytes, and they survive and integrate after xenotransplantation. Whole genome expression profiling of astrocyte differentiation was performed at several time points of differentiation, and the results indicate the importance of known regulators and identify potential novel regulators and stage-specific lineage markers.
Collapse
|
28
|
Lu HX, Yang ZQ, Jiao Q, Wang YY, Wang L, Yang PB, Chen XL, Zhang PB, Wang P, Chen MX, Lu XY, Liu Y. Low concentration of serum helps to maintain the characteristics of NSCs/NPCs on alkali-treated PHBHHx filmin vitro. Neurol Res 2014; 36:207-14. [DOI: 10.1179/1743132813y.0000000281] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
29
|
Langenfeld E, Deen M, Zachariah E, Langenfeld J. Small molecule antagonist of the bone morphogenetic protein type I receptors suppresses growth and expression of Id1 and Id3 in lung cancer cells expressing Oct4 or nestin. Mol Cancer 2013; 12:129. [PMID: 24160469 PMCID: PMC4176118 DOI: 10.1186/1476-4598-12-129] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 10/17/2013] [Indexed: 01/06/2023] Open
Abstract
Background Bone morphogenetic proteins (BMP) are embryonic morphogens that are aberrantly expressed in lung cancer. BMPs mediate cell fate decisions and self-renewal of stem cells, through transcription regulation of inhibitor of differentiation protein/DNA binding proteins (Id1-3). Inhibition of BMP signaling decreases growth and induces cell death of lung cancer cells lines by downregulating the expression of Id proteins. It is not known whether the BMP signaling cascade regulates growth and the expression of Id proteins of lung cancer cells expressing the stem cell markers Oct4 and/or nestin. Methods Lung cancer cells expressing Oct4 or nestin were isolated from lung cancer cell lines by stably transfecting the Oct4 promoter or nestin promoter expression vectors that induce expression of the green fluorescent protein reporter. Results Our studies suggest that lung cancer cells expressing Oct4 or nestin are different cell populations. Microarray and quantitative RT-PCR demonstrated that the expression of specific stem cell markers were different between isolated Oct4 and nestin cells. Both the Oct4 and nestin populations were more tumorigenic than controls but histologically they were quite different. The isolated Oct4 and nestin cells also responded differently to inhibition of BMP signaling. Blockade of BMP signaling with the BMP receptor antagonist DMH2 caused significant growth inhibition of both the Oct4 and nestin cell populations but only increased cell death in the nestin population. DMH2 also induced the expression of nestin in the Oct4 population but not in the nestin cells. We also show that BMP signaling is an important regulator of Id1 and Id3 in both the Oct4 and nestin cell populations. Furthermore, we show that NeuN is frequently expressed in NSCLC and provide evidence suggesting that Oct4 cells give rise to cancer cells expressing nestin and/or NeuN. Conclusion These studies show that although biologically different, BMP signaling is growth promoting in cancer cells expressing Oct4 or nestin. Inhibition of BMP signaling decreases expression of Id proteins and suppresses growth of cancer cells expressing Oct4 or Nestin. Small molecule antagonists of the BMP type I receptors represent potential novel drugs to target the population of cancer cells expressing stem cell markers.
Collapse
Affiliation(s)
| | | | | | - John Langenfeld
- Division of Thoracic Surgery, Rutgers-Robert Wood Johnson Medical School, One Robert Wood Johnson Place, P,O, Box 19, New Brunswick, NJ 08903-0019, USA.
| |
Collapse
|
30
|
Wang K, Long Q, Jia C, Liu Y, Yi X, Yang H, Fei Z, Liu W. Over-expression of Mash1 improves the GABAergic differentiation of bone marrow mesenchymal stem cells in vitro. Brain Res Bull 2013; 99:84-94. [PMID: 24144723 DOI: 10.1016/j.brainresbull.2013.10.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 10/08/2013] [Accepted: 10/10/2013] [Indexed: 01/11/2023]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) have been shown to be a promising cell type for the study of neuronal differentiation; however, few attempts had been made to differentiate these cells into inhibitory gamma-aminobutyric acid (GABA)ergic neurons. In this study, we over-expressed mammalian achaete-scute homologue-1 (Mash1), a basic helix-loop-helix (bHLH) transcription factor, in Sprague-Dawley rat BMSCs via lentiviral vectors, and then induced neuronal differentiation of these cells using conditioned medium. Our Western blot results show that, under conditions of differentiation, Mash1-overexpressing BMSCs exhibit an increased expression of neuronal markers and a greater degree of neuronal morphology compared to control, non-Mash1-overexpressing cells. Using immunocytochemistry, we observed increased expression of glutamic acid decarboxylase 67 (GAD67), as well as neuron-specific nuclear protein (NeuN) and β3-tubulin, in Mash1-overexpressing BMSCs compared to control cells. Moreover, we also found the differentiated cells showed representative traces of action potentials in electrophysiological characterization. In conclusion, our study demonstrated that over-expression of Mash1 can improve GABAergic differentiation of BMSCs in vitro.
Collapse
Affiliation(s)
- Kai Wang
- Department of Neurosurgery, Xijing Institute of Clinical Neuroscience, Fourth Military Medical University, No.17 Chang-le West Road, Xi'an 710032, China; Department of Neurosurgery, Qingdao 401 Hospital of PLA, No. 22 Minjiang Road, Qingdao 266071, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Buda R, Vannini F, Cavallo M, Baldassarri M, Luciani D, Mazzotti A, Pungetti C, Olivieri A, Giannini S. One-step arthroscopic technique for the treatment of osteochondral lesions of the knee with bone-marrow-derived cells: three years results. Musculoskelet Surg 2013; 97:145-151. [PMID: 23420394 DOI: 10.1007/s12306-013-0242-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 01/11/2013] [Indexed: 06/01/2023]
Abstract
Osteochondral lesions of the knee (OLK) are a common cause of knee pain and associated diseases. A new bone-marrow-derived mesenchymal stem cells technique has been developed for the treatment of OLK. 30 patients with OLK underwent arthroscopic one-step procedure. The bone marrow was harvested from the patients' posterior iliac crest and arthroscopically implanted with a scaffold into the lesion site. Clinical inspection and MRI were performed. Mean International Knee Documentation Committee (IKDC) score before surgery was 29.9 ± 13.2 and 85.4 ± 4.2 at 29 ± 4.1 months (p < 0.0005), while Knee injury and Osteoarthritis Outcome Score (KOOS) before surgery was 35.1 ± 11.9 and 87.3 ± 7.3 at 29 ± 4.1 months (p < 0.0005). Control MRI and bioptic samples showed an osteochondral regeneration of the lesion site. The one-step technique appears to be a good and reliable option for treatment of OLK at three years of follow-up. Level of evidence Case series, Level IV.
Collapse
Affiliation(s)
- Roberto Buda
- Clinical Orthopaedic and Traumatology Unit I, Rizzoli Orthopaedic Institute, via G.C. Pupilli n.1, Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bonner JF, Haas CJ, Fischer I. Preparation of neural stem cells and progenitors: neuronal production and grafting applications. Methods Mol Biol 2013; 1078:65-88. [PMID: 23975822 DOI: 10.1007/978-1-62703-640-5_7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neural stem cells (NSC) are not only a valuable tool for the study of neural development and function, but an integral component in the development of transplantation strategies for neural disease. NSC can be used to study how neurons acquire distinct phenotypes and how the reciprocal interactions between neurons and glia in the developing nervous system shape the structure and function of the central nervous system (CNS). In addition, neurons prepared from NSC can be used to elucidate the molecular basis of neurological disorders as well as potential treatments. Although NSC can be derived from different species and many sources, including embryonic stem cells, induced pluripotent stem cells, adult CNS, and direct reprogramming of non-neural cells, isolating primary NSC directly from rat fetal tissue is the most common technique for preparation and study of neurons with a wealth of data available for comparison. Regardless of the source material, similar techniques are used to maintain NSC in culture and to differentiate NSC toward mature neural lineages. This chapter will describe specific methods for isolating multipotent NSC and neural precursor cells (NPC) from embryonic rat CNS tissue (mostly spinal cord). In particular, NPC can be separated into neuronal and glial restricted precursors (NRP and GRP, respectively) and used to reliably produce neurons or glial cells both in vitro and following transplantation into the adult CNS. This chapter will describe in detail the methods required for the isolation, propagation, storage, and differentiation of NSC and NPC isolated from rat spinal cords for subsequent in vitro or in vivo studies.
Collapse
Affiliation(s)
- Joseph F Bonner
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, USA
| | | | | |
Collapse
|
33
|
Lu X, Wang L, Yang Z, Lu H. Strategies of polyhydroxyalkanoates modification for the medical application in neural regeneration/nerve tissue engineering. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abb.2013.46097] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
34
|
Yang Z, Qiao H, Sun Z, Li X. Effect of BDNF-plasma-collagen matrix controlled delivery system on the behavior of adult rats neural stem cells. J Biomed Mater Res A 2012; 101:599-606. [PMID: 23090850 DOI: 10.1002/jbm.a.34331] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 06/24/2012] [Accepted: 06/26/2012] [Indexed: 12/14/2022]
Abstract
The neurogenesis amount in central nervous system (CNS) stimulated by the injury or diseases is so small that neural stem cells (NSCs) cannot specifically differentiate into the ideal phenotypes to repair the injured CNS. The transplanted exogenous NSCs also have such problems as poor survival and insufficient neuronal differentiation. In this study, the behavior of NSCs from the spinal cord of adult rats was compared at the neurosphere level after the respective addition of the brain-derived neurotrophic factor (BDNF) daily, the BDNF-loaded plasma-collagen matrix, the plasma-collagen matrix alone, or the defined medium alone. The results suggested that the BDNF, either in the control release form or in the soluble form, initiated NSCs proliferation and differentiation by activating receptors Trk B and p75NTR. BDNF also increased the differentiation percentage of adult NSCs into neurons and supported the long-term cell survival and growth. The BDNF was stably released by the plasma-collagen matrix for up to 21 days. The plasma-collagen matrix alone showed its biocompatibility with cells by facilitating the adhesion, survival, and differentiation of NSCs. The NSCs in the defined medium alone group showed poor survival and a very low level of neuronal differentiation and proliferation abilities than above three groups. This study suggested that the BDNF-loaded plasma-collagen matrix may provide a promising means to resolve either the poor survival and insufficient neuronal differentiation of transplanted exogenous NSCs, or stimulating the intrinsic NSCs to proliferate and differentiate into neurons so as to repair the injured adult CNS.
Collapse
Affiliation(s)
- Zhaoyang Yang
- Beijing Institute for Neuroscience, Capital Medical University, Beijing 100069, China
| | | | | | | |
Collapse
|
35
|
Fainstein N, Cohen ME, Ben-Hur T. Time associated decline in neurotrophic properties of neural stem cell grafts render them dependent on brain region-specific environmental support. Neurobiol Dis 2012; 49:41-8. [PMID: 22910454 DOI: 10.1016/j.nbd.2012.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 07/08/2012] [Accepted: 08/07/2012] [Indexed: 01/12/2023] Open
Abstract
Fetal neural stem/precursor cells (NPCs) possess powerful neurotrophic properties by which they can facilitate self repair processes in the adult central nervous system. The therapeutic value of NPC therapy in neurodegenerative diseases is critically dependent on their long term survival and enduring functional properties. An important aspect of NPC neurotrophic properties is their ability to support their own survival independent of any exogenous growth factor. Here, we examined whether NPCs survive and maintain their properties for extended periods of time, or become dependent on environmental support. Two months following transplantation to naïve brains, large grafts were detected in the ventricles and hippocampus, but only little survival was evident in the striatum. To point at possible regional characteristics which underlie the differential survival of NPC grafts we performed several manipulations of the brain environment. Acute neurotoxic injury with 6-hydroxydopamine induced a 3-fold increase in striatal graft survival, associated with induction of nestin, CD31, β1-integrin, GFAP and cycling cells. Disruption of the extracellular matrix structure of this reactive niche by continuous blockage of host striatum β1-integrin caused 73% reduction in graft survival. In the hippocampus, NPC graft survival did not correspond to β1-integrin and CD31 expression. This suggests that hippocampal environmental support to graft survival rely on different mechanisms than in the reactive striatum. In correlation with in vivo findings, long term cultured neural precursors exhibited an increase in apoptotic cells and dramatic decline in neurotrophic effects, as indicated by two in vitro functional assays. We conclude that long-term changes in transplanted NPC properties render them dependent on region specific environmental support. The major extracellular matrix protein β1-integrin is essential for providing tissue support for graft survival in the striatum. The neurotrophic properties of transplanted neural stem cells are limited in time, representing a shortcoming which should be taken into consideration when developing clinical transplantation protocols for chronic neurological disorders.
Collapse
Affiliation(s)
- Nina Fainstein
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, P.O. Box 12,000, Jerusalem 91120, Israel
| | - Mikhal E Cohen
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, P.O. Box 12,000, Jerusalem 91120, Israel
| | - Tamir Ben-Hur
- Department of Neurology, The Agnes Ginges Center for Human Neurogenetics, Hadassah - Hebrew University Medical Center, P.O. Box 12,000, Jerusalem 91120, Israel.
| |
Collapse
|
36
|
Jin Y, Sura K, Fischer I. Differential effects of distinct central nervous system regions on cell migration and axonal extension of neural precursor transplants. J Neurosci Res 2012; 90:2065-73. [PMID: 22740505 DOI: 10.1002/jnr.23099] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 04/11/2012] [Accepted: 05/16/2012] [Indexed: 11/06/2022]
Abstract
Transplantation of neural precursor cells (NPCs) is a promising therapeutic strategy in CNS injury. However, the adult CNS lacks instructive signals present during development and, depending on the region and type of transplant, may be inhibitory for neuron generation and axonal growth. We examined the effects of the white matter in different regions of the adult CNS on the properties of NPC transplants with respect to cell survival, differentiation, migration, and axonal growth. NPCs were prepared from day 13.5 embryonic spinal cord of transgenic rats that express the human placental alkaline phosphatase (AP) reporter. These NPCs were injected unilaterally into the cervical spinal cord white matter and into the corpus callosum of adult rats and were analyzed immunohistochemically 2 weeks later. NPCs survived in both regions and differentiated into astrocytes, oligodendrocytes, and neurons, with no apparent differences in survival or phenotypic composition. However, in the spinal cord white matter, graft-derived cells, identified as precursors and glial cells, migrated from the injection site rostrally and caudally, whereas, in the corpus callosum, graft-derived cells did not migrate and remained at the injection site. Importantly, graft-derived neurons extended axons from the grafting site along the corpus callosum past the midline, entering into the contralateral side of the corpus callosum. These results demonstrate dramatic differences between white matter regions in the spinal cord and brain with respect to cell migration and axonal growth and underscore the importance of considering the effects of the local CNS environment in the design of effective transplantation strategies.
Collapse
Affiliation(s)
- Ying Jin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, USA.
| | | | | |
Collapse
|
37
|
Phillips AW, Falahati S, DeSilva R, Shats I, Marx J, Arauz E, Kerr DA, Rothstein JD, Johnston MV, Fatemi A. Derivation of glial restricted precursors from E13 mice. J Vis Exp 2012:3462. [PMID: 22760029 PMCID: PMC3399460 DOI: 10.3791/3462] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
This is a protocol for derivation of glial restricted precursor (GRP) cells from the spinal cord of E13 mouse fetuses. These cells are early precursors within the oligodendrocytic cell lineage. Recently, these cells have been studied as potential source for restorative therapies in white matter diseases. Periventricular leukomalacia (PVL) is the leading cause of non-genetic white matter disease in childhood and affects up to 50% of extremely premature infants. The data suggest a heightened susceptibility of the developing brain to hypoxia-ischemia, oxidative stress and excitotoxicity that selectively targets nascent white matter. Glial restricted precursors (GRP), oligodendrocyte progenitor cells (OPC) and immature oligodendrocytes (preOL) seem to be key players in the development of PVL and are the subject of continuing studies. Furthermore, previous studies have identified a subset of CNS tissue that has increased susceptibility to glutamate excitotoxicity as well as a developmental pattern to this susceptibility. Our laboratory is currently investigating the role of oligodendrocyte progenitors in PVL and use cells at the GRP stage of development. We utilize these derived GRP cells in several experimental paradigms to test their response to select stresses consistent with PVL. GRP cells can be manipulated in vitro into OPCs and preOL for transplantation experiments with mouse PVL models and in vitro models of PVL-like insults including hypoxia-ischemia. By using cultured cells and in vitro studies there would be reduced variability between experiments which facilitates interpretation of the data. Cultured cells also allows for enrichment of the GRP population while minimizing the impact of contaminating cells of non-GRP phenotype.
Collapse
Affiliation(s)
- André W Phillips
- Hugo W. Moser Research Institute at Kennedy Krieger, Johns Hopkins University.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Ju PJ, Liu R, Yang HJ, Xia YY, Feng ZW. Clonal analysis for elucidating the lineage potential of embryonic NG2+ cells. Cytotherapy 2012; 14:608-20. [DOI: 10.3109/14653249.2011.651528] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
39
|
Yazdani SO, Pedram M, Hafizi M, Kabiri M, Soleimani M, Dehghan MM, Jahanzad I, Gheisari Y, Hashemi SM. A comparison between neurally induced bone marrow derived mesenchymal stem cells and olfactory ensheathing glial cells to repair spinal cord injuries in rat. Tissue Cell 2012; 44:205-13. [PMID: 22551686 DOI: 10.1016/j.tice.2012.03.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2011] [Revised: 03/22/2012] [Accepted: 03/22/2012] [Indexed: 02/08/2023]
Abstract
Cell therapy has proven to be a highly promising method in clinical applications, raising so much hope for the treatment of injured tissues with low, if any, self regeneration potential such as central and peripheral nervous system. Neurally induced bone marrow derived mesenchymal stem cells (NIMSCs) as well as olfactory ensheathing cells (OECs) were transplanted in a rat model of sub-acute spinal cord injury and the behavioral and histological analyses were conducted. A balloon-compression technique was used to produce an injury at T8-T9 level of spinal cord. After a week post injury, rats were injected with either NIMSCs or OECs at the center of developing lesion cavity, 3mm cranial and 3mm caudal to the cavity. Weekly behavioral assessment using BBB score was done over five-week period post transplantation and finally histological assessment was performed to locate labeled cells in the tissue in order to evaluate the reduction of cavity formation and axonal regeneration. Evaluation of locomotor performance showed significant behavioral improvement in NIMSC group over OEC and control groups. The histological analyses revealed the presence of transplanted cells in the spinal cord parenchyma. Volume of injured area that was occupied with syrinx cavity in NIMSC group was significantly less than control group. In addition, meanwhile neurofilament-positive axons significantly showed higher expression in rats receiving NIMSC compared to the other two groups. In conclusion NIMSC caused both behavioral and histological improvement that potentially makes them a promising candidate for cell therapy approaches of spinal cord injuries.
Collapse
Affiliation(s)
- Saeed Oraee Yazdani
- Students' Scientific Research Center of Tehran University of Medical Sciences-SSRC, Tehran University of Medical Sciences, Medical Faculty, Tehran, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
The roles of neuronal and glial precursors in overcoming chondroitin sulfate proteoglycan inhibition. Exp Neurol 2012; 235:627-37. [PMID: 22498104 DOI: 10.1016/j.expneurol.2012.03.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 03/26/2012] [Indexed: 11/23/2022]
Abstract
The extension of axons through the major inhibitory component of the glial scar, chondroitin sulfate proteoglycans (CSPGs), remains a key obstacle for regeneration following spinal cord injury (SCI). We have previously shown that transplants composed of neuronal and glial restricted precursors (NRP and GRP respectively) promote regeneration and connectivity in the injured spinal cord (Bonner et al., 2010, 2011), however, little is known about the properties of these precursors at a cellular level. We now report that NRP-derived neurons, in contrast to dorsal root ganglion (DRG) neurons, have the ability to extend axons and cross over from a permissive substratum (laminin) onto inhibitory CSPG in vitro. Growth cones of neurons derived from NRP, compared to DRG, exhibit significantly lower levels of the CSPG receptors protein tyrosine phosphatase sigma (PTPσ) and leukocyte common antigen-related phosphatase (LAR). GRP-conditioned medium prepared from the same cell densities did not affect the response of primary sensory neurons to CSPG confirming that the ability of NRP-derived neurons to cross onto CSPG is determined intrinsically. However, GRP-conditioned medium collected from high density cultures increased the probability of DRG axons to cross from LN onto CSPG and increased the length of DRG axons extending on CSPG. Collectively, these results suggest that (1) neurons derived from NRPs are intrinsically insensitive to CSPGs due to low levels of receptor expression, and (2) high levels of factors secreted by GRP can reduce the inhibitory effects of CSPG and promote axonal growth. These observations provide mechanistic insights into the specific roles of NRPs and GRPs in promoting regeneration and repair following SCI.
Collapse
|
41
|
Lu HX, Hao ZM, Jiao Q, Xie WL, Zhang JF, Lu YF, Cai M, Wang YY, Yang ZQ, Parker T, Liu Y. Neurotrophin-3 gene transduction of mouse neural stem cells promotes proliferation and neuronal differentiation in organotypic hippocampal slice cultures. Med Sci Monit 2012; 17:BR305-311. [PMID: 22037732 PMCID: PMC3539508 DOI: 10.12659/msm.882039] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The transplantation of neural stem cells (NSCs) has been accepted as a promising therapeutic strategy for central nervous system disorders. However, the beneficial effect of NSC transplantation upon functional recovery is limited due to the unfavorable microenvironment (niche) at the site of trauma or degenerative disease in the brain. Combination of transplantation of NSCs with neurotrophins may overcome the hurdles of impaired cell survival and neuronal differentiation. MATERIAL/METHODS In the current study, the neurotrophin-3 (NT-3) gene was transduced into cultured mouse embryonic cortical NSCs via an AAV vector (NSC-NT-3). The effect of NT-3 over-expression on cell proliferation and differentiation in NSCs was observed by immunohistochemistry, cell culture and organotypic hippocampal slice cultures.<br /> RESULTS The characteristics of self-renewal and multiple differentiation of NSCs were well-preserved. Cells in the NSC-NT-3 group proliferated faster and differentiated into more β-tubulin III-positive neurons compared to the control group in vitro. Furthermore, cells in the NSC-NT-3 group survived in a significantly higher percentage and undertook neuronal differentiation preferably in organotypic hippocampal slice cultures. CONCLUSIONS Our results suggest that the transduction of NT-3 into NSCs could effectively promote NSCs survival, proliferation, and neuronal differentiation in vitro without change of the stemness of NSCs. This work also offers evidence to better understand the safety and efficiency of combined treatment with NT-3 and NSCs for the central nervous system disorders.
Collapse
Affiliation(s)
- Hai-xia Lu
- Institute of Neurobiology, Environment and Genes Related to Diseases Key Laboratory of Education Ministry, Xi'an Jiaotong University College of Medicine, Xi'an, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mitsui T, Neuhuber B, Fischer I. Acute administration of AMPA/Kainate blocker combined with delayed transplantation of neural precursors improves lower urinary tract function in spinal injured rats. Brain Res 2011; 1418:23-31. [PMID: 21937028 DOI: 10.1016/j.brainres.2011.08.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Revised: 08/10/2011] [Accepted: 08/10/2011] [Indexed: 11/28/2022]
Abstract
To evaluate bladder function recovery after spinal cord injury (SCI) in response to a combination treatment of an acutely administered AMPA/kainate receptor antagonist and delayed transplantation of neuronal precursors. Female rats received a contusion injury at T8/9. The AMPA/kainate receptor antagonist NBQX was directly administered into the lesion site immediately after injury. Nine days post-injury, NRP/GRP were delivered into the lesion site. Controls received NRP/GRP grafts only or no treatment (OP-Controls). Animals underwent bladder function testing during the course of the experiment and at the endpoint. Motor function was evaluated as well. After sacrifice, histological analysis of lesion site and lumbosacral spinal cord regions was performed. Rats receiving the combined treatment (NBQX&NRP/GRP) had voided volumes/micturition resembling that of normal animals and showed greater improvement of urodynamic parameters, compared to NRP/GRP alone or OP-Controls. Similarly, NBQX&NRP/GRP induced more spouting, regeneration or sparing of descending projections to the lumbosacral cord. The density of primary afferent projections at the lumbosacral spinal cord in rats with combined treatments was similar to that of NRP/GRP alone with decreased sprouting of primary afferents in lumbosacral cord, compared to OP-Control. Immunohistochemical evaluation revealed that the combined treatment reduced the size of the lesion to a greater extent than NRP/GRP alone or OP-Controls. NRP/GRP with and without NBQX produced a significant recovery of hindlimb compared to OP-Controls. In conclusion, transplants of NRP/GRP combined with NBQX promote recovery of micturition function following spinal cord injury, likely through increased neuroprotection.
Collapse
Affiliation(s)
- Takahiko Mitsui
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA.
| | | | | |
Collapse
|
43
|
Jin Y, Ketschek A, Jiang Z, Smith G, Fischer I. Chondroitinase activity can be transduced by a lentiviral vector in vitro and in vivo. J Neurosci Methods 2011; 199:208-13. [PMID: 21600922 DOI: 10.1016/j.jneumeth.2011.05.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 04/18/2011] [Accepted: 05/04/2011] [Indexed: 12/11/2022]
Abstract
The bacterial enzyme chondroitinase ABC (ChABC), which cleaves chondroitin sulfate glycosaminoglycan chains, can degrade inhibitory scar tissue formed following spinal cord injury, thereby promoting axonal growth and regeneration. However, delivering the active enzyme for prolonged periods presents practical limitations. To overcome these problems, we prepared a lentiviral vector (LV) encoding chondroitinase AC (Chase) together with the green fluorescent protein (GFP) reporter (Chase/LV) and demonstrated its expression and enzymatic activity in vitro and in vivo. Neural precursor cells infected with Chase/LV expressed the GFP reporter at levels that increased dramatically with time in culture. Enzymatic activity from the supernatant of the infected cells was demonstrated by dot blot assay using an antibody that recognizes the digested form of CSPG and was compared with the bacterial ChABC enzyme. Chick DRG cultures plated adjacent to the CSPG border and incubated with supernatant from Chase/LV-infected cells showed neurites growing into the CSPG area, a response similar to that after treatment with ChABC. In contrast, in control cultures, the neurites turned to avoid the inhibitory CSPG interface. Degradation of CSPG in these cultures was confirmed by specific CSPG antibodies. A single injection of Chase/LV into the spinal cord resulted in sustained secretion of the enzyme, whose activity was detected for 8 weeks by expression of GFP and evidence of the digested form of CSPG. This study demonstrates the efficacy of the Chase/LV vector and its potential as a therapeutic tool to reduce scar inhibition and promote axonal growth and repair following central nervous system injury.
Collapse
Affiliation(s)
- Ying Jin
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, United States.
| | | | | | | | | |
Collapse
|
44
|
Lepore AC, O'Donnell J, Bonner JF, Paul C, Miller ME, Rauck B, Kushner RA, Rothstein JD, Fischer I, Maragakis NJ. Spatial and temporal changes in promoter activity of the astrocyte glutamate transporter GLT1 following traumatic spinal cord injury. J Neurosci Res 2011; 89:1001-17. [PMID: 21488085 DOI: 10.1002/jnr.22624] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 12/20/2010] [Accepted: 01/19/2011] [Indexed: 12/21/2022]
Abstract
After traumatic spinal cord injury (SCI), there is an opportunity for preserving function by attenuating secondary cell loss. Astrocytes play crucial roles in the adult CNS and are responsible for the vast majority of glutamate buffering, potentially preventing excitotoxic loss of neurons and oligodendrocytes. We examined spatial and temporal changes in gene expression of the major astrocyte glutamate transporter GLT1 following moderate thoracic contusion SCI using transgenic BAC-GLT1-eGFP promoter reporter mice. In dorsal column white matter, total intensity of GLT1-eGFP expression per region was significantly reduced following SCI at both lesion epicenter and at rostral and caudal areas where no tissue loss occurred. This regional decrease in GLT1 expression was due to significant loss of GLT1-eGFP(+) cells, partially accounted for by apoptosis of eGFP(+) /GFAP(+) astrocytes in both white and gray matter. There were also decreased numbers of GLT1-eGFP-expressing cells in multiple gray matter regions following injury; nevertheless, there was sustained or even increased regional GLT1-eGFP expression in gray matter as a result of up-regulation in astrocytes that continued to express GLT1-eGFP. Although there were increased numbers of GFAP(+) cells both at the lesion site and in surrounding intact spinal cord following SCI, the majority of proliferating Ki67(+) /GFAP(+) astrocytes did not express GLT1-eGFP. These findings demonstrate that spatial and temporal alterations in GLT1 expression observed after SCI result from both astrocyte death and gene expression changes in surviving astrocytes. Results also suggest that following SCI a significant portion of astrocytes lacks GLT1 expression, possibly compromising the important role of astrocytes in glutamate homeostasis.
Collapse
Affiliation(s)
- Angelo C Lepore
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Dagci T, Sengul G, Keser A, Onal A. NADPH-d and Fos reactivity in the rat spinal cord following experimental spinal cord injury and embryonic neural stem cell transplantation. Life Sci 2011; 88:746-52. [PMID: 21376061 DOI: 10.1016/j.lfs.2011.02.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 12/28/2010] [Accepted: 02/03/2011] [Indexed: 12/25/2022]
Abstract
AIMS The aim of this study is to determine the role of nitric oxide (NO) in neuropathic pain and the effect of embryonic neural stem cell (ENSC) transplantation on NO content in rat spinal cord neurons following spinal cord injury (SCI). MAIN METHODS Ninety adult male Sprague-Dawley rats were divided into 3 groups (n=30, each): control (laminectomy), SCI (hemisection at T12-T13 segments) and SCI+ENSC. Each group was further divided into sub-groups (n=5 each) based on the treatment substance (L-NAME, 75 mg/kg/i.p.; L-arginine, 225 mg/kg/i.p.; physiological saline, SF) and duration (2h for acute and 28 days for chronic groups). Pain was assessed by tail flick and Randall-Selitto tests. Fos immunohistochemistry and NADPH-d histochemistry were performed in segments 2 cm rostral and caudal to SCI. KEY FINDINGS Tail-flick latency time increased in both acute and chronic L-NAME groups and increased in acute and decreased in chronic L-arginine groups. The number of Fos (+) neurons decreased in acute and chronic L-NAME and decreased in acute L-arginine groups. Following ENSC, Fos (+) neurons did not change in acute L-NAME but decreased in the chronic L-NAME groups, and decreased in both acute and chronic L-arginine groups. NADPH-d (+) neurons decreased in acute L-NAME and increased in L-arginine groups with and without ENSC transplantation. SIGNIFICANCE This study confirms the role of NO in neuropathic pain and shows an improvement following ENSC transplantation in the acute phase, observed as a decrease in Fos(+) and NADPH-d (+) neurons in spinal cord segments rostral and caudal to injury.
Collapse
Affiliation(s)
- Taner Dagci
- Department of Physiology, Ege University, School of Medicine, Bornova, Izmir, Turkey
| | | | | | | |
Collapse
|
46
|
Abstract
Amyotrophic lateral sclerosis (ALS), a disorder that affects 30,000 individuals in the USA alone, is characterized by relatively rapid degeneration of upper and lower motor neurons, with death normally occurring 2-5 years following diagnosis due to respiratory paralysis. Transplantation of various classes of neural precursor cells (NPCs) is a promising therapeutic strategy for the treatment of traumatic CNS injury and neurodegeneration, including ALS, because of the ability to replace lost or dysfunctional CNS cell types, provide neuroprotection, and deliver gene factors of interest. In order to target cellular therapy to diaphragmatic dysfunction in models of ALS, NPCs can be transplanted specifically into the cervical spinal cord ventral gray matter of both SOD1G93A rats and mice. The SOD1G93A rats and mice are currently the most well-studied animal model of the disease.
Collapse
Affiliation(s)
- Angelo C Lepore
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
47
|
See J, Bonner J, Neuhuber B, Fischer I. Neurite outgrowth of neural progenitors in presence of inhibitory proteoglycans. J Neurotrauma 2010; 27:951-7. [PMID: 20102265 DOI: 10.1089/neu.2009.1158] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Attempts to promote host regeneration after spinal cord injury (SCI) have often resulted in poor axon extension due to formation of a glial scar, which creates a dense physical barrier around the injury and contains molecules that inhibit regeneration and repair of adult injured axons. Previous studies have shown that, while transplants of multipotent neural stem cells (NSC) integrate poorly in the injury site, the use of neuronal-restricted precursor cells (NRP) together with glial-restricted precursor cells (GRP) allow differentiation and integration of neurons, possibly because NRP are able to overcome chondroitin sulfate proteoglycan (CSPG) inhibition. To investigate this possibility, we grew mixed cultures of NRP/GRP on CSPG at inhibitory concentrations, using embryonic hippocampal cultures as controls. We found that NRP/GRP grown on CSPG survive and differentiate into neurons with no significant changes in neurite length, relative to growth on control polylysine substrate, and in contrast to a significant inhibition of axon growth in hippocampal cultures grown on CSPG-coated substrate. There was, however, a significant decrease in neurite number and branching in both cultures, indicating that CSPG also has important effects on neuronal morphology. These data suggest that embryonic neurons supported by glial cells derived from NRP/GRP transplants are less sensitive to inhibitory effects of CSPG in the glial scar, and are thus an appropriate source for neuronal cell replacement and reconnection of damaged circuits after SCI.
Collapse
Affiliation(s)
- Jill See
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania 19129, USA
| | | | | | | |
Collapse
|
48
|
Buda R, Vannini F, Cavallo M, Grigolo B, Cenacchi A, Giannini S. Osteochondral lesions of the knee: a new one-step repair technique with bone-marrow-derived cells. J Bone Joint Surg Am 2010; 92 Suppl 2:2-11. [PMID: 21123588 DOI: 10.2106/jbjs.j.00813] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Roberto Buda
- II Clinic of Orthopaedics and Traumatology, Rizzoli Orthopaedic Institute, Bologna, Italy
| | | | | | | | | | | |
Collapse
|
49
|
Effects of salvianolic acid B on proliferation, neurite outgrowth and differentiation of neural stem cells derived from the cerebral cortex of embryonic mice. SCIENCE CHINA-LIFE SCIENCES 2010; 53:653-62. [DOI: 10.1007/s11427-010-3106-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2008] [Accepted: 06/06/2009] [Indexed: 01/03/2023]
|
50
|
Bonner JF, Blesch A, Neuhuber B, Fischer I. Promoting directional axon growth from neural progenitors grafted into the injured spinal cord. J Neurosci Res 2010; 88:1182-92. [PMID: 19908250 DOI: 10.1002/jnr.22288] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Spinal cord injury (SCI) is a devastating condition characterized by disruption of axonal connections, failure of axonal regeneration, and loss of motor and sensory function. The therapeutic promise of neural stem cells has been focused on cell replacement, but many obstacles remain in obtaining neuronal integration following transplantation into the injured CNS. This study investigated the neurotransmitter identity and axonal growth potential of neural progenitors following grafting into adult rats with a dorsal column lesion. We found that using a combination of neuronal and glial restricted progenitors (NRP and GRP) produced graft-derived glutamatergic and GABAergic neurons within the injury site, with minimal axonal extension. Administration of brain-derived neurotrophic factor (BDNF) with the graft promoted modest axonal growth from grafted cells. In contrast, injecting a lentiviral vector expressing BDNF rostral into the injured area generated a neurotrophin gradient and promoted directional growth of axons for up to 9 mm. Animals injected with BDNF lentivirus (at 2.5 and 5.0 mm) showed significantly more axons and significantly longer axons than control animals injected with GFP lentivirus. However, only the 5.0-mm-BDNF group showed a preference for extension in the rostral direction. We concluded that NRP/GRP grafts can be used to produce excitatory and inhibitory neurons, and neurotrophin gradients can guide axonal growth from graft-derived neurons toward putative targets. Together they can serve as a building block for neuronal cell replacement of local circuits and formation of neuronal relays.
Collapse
Affiliation(s)
- Joseph F Bonner
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | | | | | | |
Collapse
|