1
|
Kamatani T, Kimura R, Ikeda S, Inoue M, Seino KI. iPSCs engrafted in allogeneic hosts without immunosuppression induce donor-specific tolerance to secondary allografts. Proc Natl Acad Sci U S A 2025; 122:e2413398122. [PMID: 40073064 PMCID: PMC11929385 DOI: 10.1073/pnas.2413398122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Currently, most cell or tissue transplantations using induced pluripotent stem cells (iPSCs) are anticipated to involve allogeneic iPSCs. However, the immunological properties of iPSCs in an allogeneic setting are not well understood. We previously established a mouse transplantation model of MHC-compatible/minor antigen-mismatched combinations, assuming a hypoimmunogenic iPSC-setting. Here, we found that iPSCs subcutaneously inoculated into MHC-compatible allogeneic host mice resisted rejection and formed teratomas without immunosuppressant administration. Notably, when skin grafts were transplanted onto hosts more than 40 d after the initial iPSCs inoculation, only the skin of the same strain as the initial iPSCs was engrafted. Therefore, donor-specific immune tolerance was induced by a single iPSC inoculation. Diverse analyses, including single-cell RNA-sequencing after transplantation, revealed an increase in regulatory T cell (Treg) population, particularly CD25+ CD103+ effector Tregs within the teratoma and skin grafts. The removal of CD25+ or Foxp3+ cells suppressed the increase in effector Tregs and disrupted graft acceptance, indicating the importance of these cells in the establishment of immune tolerance. Within the teratoma, we observed an increase in TGF-β2 levels, suggesting an association with the increase in effector Tregs. Our results provide important insights for future applications of allogeneic iPSC-based cell or tissue transplantation.
Collapse
Affiliation(s)
- Tomoki Kamatani
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido060-0815, Japan
| | - Reiko Kimura
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido060-0815, Japan
| | | | - Makoto Inoue
- Sumitomo Pharma, Co., Ltd., Osaka541-0045, Japan
| | - Ken-ichiro Seino
- Division of Immunobiology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido060-0815, Japan
| |
Collapse
|
2
|
Olaya AMS, Almeida FM, Martinez AMB, Marques SA. Treatment of spinal cord injury with biomaterials and stem cell therapy in non-human primates and humans. Neural Regen Res 2025; 20:343-353. [PMID: 38819038 PMCID: PMC11317961 DOI: 10.4103/nrr.nrr-d-23-01752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/27/2024] [Accepted: 03/27/2024] [Indexed: 06/01/2024] Open
Abstract
Spinal cord injury results in the loss of sensory, motor, and autonomic functions, which almost always produces permanent physical disability. Thus, in the search for more effective treatments than those already applied for years, which are not entirely efficient, researches have been able to demonstrate the potential of biological strategies using biomaterials to tissue manufacturing through bioengineering and stem cell therapy as a neuroregenerative approach, seeking to promote neuronal recovery after spinal cord injury. Each of these strategies has been developed and meticulously evaluated in several animal models with the aim of analyzing the potential of interventions for neuronal repair and, consequently, boosting functional recovery. Although the majority of experimental research has been conducted in rodents, there is increasing recognition of the importance, and need, of evaluating the safety and efficacy of these interventions in non-human primates before moving to clinical trials involving therapies potentially promising in humans. This article is a literature review from databases (PubMed, Science Direct, Elsevier, Scielo, Redalyc, Cochrane, and NCBI) from 10 years ago to date, using keywords (spinal cord injury, cell therapy, non-human primates, humans, and bioengineering in spinal cord injury). From 110 retrieved articles, after two selection rounds based on inclusion and exclusion criteria, 21 articles were analyzed. Thus, this review arises from the need to recognize the experimental therapeutic advances applied in non-human primates and even humans, aimed at deepening these strategies and identifying the advantages and influence of the results on extrapolation for clinical applicability in humans.
Collapse
Affiliation(s)
- Ana Milena Silva Olaya
- PhD Program in Pathological Anatomy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Ana Maria Blanco Martinez
- Graduate Program in Pathological Anatomy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suelen Adriani Marques
- Graduate Program in Pathological Anatomy (PPGAP/UFRJ), Department of Neurobiology/Institute of Biology, Campus do Gragoatá, Niterói, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
McKenna SL, Ehsanian R, Liu CY, Steinberg GK, Jones L, Lebkowski JS, Wirth E, Fessler RG. Ten-year safety of pluripotent stem cell transplantation in acute thoracic spinal cord injury. J Neurosurg Spine 2022; 37:321-330. [PMID: 35364569 DOI: 10.3171/2021.12.spine21622] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 12/20/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The purpose of this study was to evaluate the safety of oligodendrocyte progenitor cells (LCTOPC1) derived from human pluripotent stem cells administered between 7 and 14 days postinjury to patients with T3 to T11 neurologically complete spinal cord injury (SCI). The rationale for this first-in-human trial was based on evidence that administration of LCTOPC1 supports survival and potential repair of key cellular components and architecture at the SCI site. METHODS This study was a multisite, open-label, single-arm interventional clinical trial. Participants (n = 5) received a single intraparenchymal injection of 2 × 106 LCTOPC1 caudal to the epicenter of injury using a syringe positioning device. Immunosuppression with tacrolimus was administered for a total of 60 days. Participants were followed with annual in-person examinations and MRI for 5 years at the time of this report and will be followed with annual telephone questionnaires for 6 to 15 years postinjection. The primary endpoint was safety, as measured by the frequency and severity of adverse events related to the LCTOPC1 injection, the injection procedure, and/or the concomitant immunosuppression administered. The secondary endpoint was neurological function as measured by sensory scores and lower-extremity motor scores as measured by the International Standards for Neurological Classification of Spinal Cord Injury examinations. RESULTS No unanticipated serious adverse events related to LCTOPC1 have been reported with 98% follow-up of participants (49 of 50 annual visits) through the first 10 years of the clinical trial. There was no evidence of neurological decline, enlarging masses, further spinal cord damage, or syrinx formation. MRI results during the long-term follow-up period in patients administered LCTOPC1 cells showed that 80% of patients demonstrated T2 signal changes consistent with the formation of a tissue matrix at the injury site. CONCLUSIONS This study provides crucial first-in-human safety data supporting the pursuit of future human embryonic stem cell-derived therapies. While we cannot exclude the possibility of future adverse events, the experience in this trial provides evidence that this cell type can be well tolerated by patients, with an event-free period of up to 10 years. Based on the safety profile of LCTOPC1 obtained in this study, a cervical dose escalation trial was initiated (NCT02302157).
Collapse
Affiliation(s)
- Stephen L McKenna
- 1Department of Physical Medicine and Rehabilitation, Santa Clara Valley Medical Center, San Jose, California
- 2Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Reza Ehsanian
- 3Division of Physical Medicine and Rehabilitation, Department of Orthopedics & Rehabilitation, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Charles Y Liu
- 4USC Neurorestoration Center, Los Angeles, California
- 6Rancho Los Amigos National Rehabilitation Center, Downey, California
| | - Gary K Steinberg
- 2Department of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Linda Jones
- 7Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Jane S Lebkowski
- 8Asterias Biotherapeutics, a wholly owned subsidiary of Lineage Cell Therapeutics, Carlsbad, California
- 9Regenerative Patch Technologies, LLC, Menlo Park, California
| | - Edward Wirth
- 8Asterias Biotherapeutics, a wholly owned subsidiary of Lineage Cell Therapeutics, Carlsbad, California
- 10Aspen Neuroscience, San Diego, California; and
| | - Richard G Fessler
- 11Department of Neurosurgery, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
4
|
Liu T, Zhu W, Zhang X, He C, Liu X, Xin Q, Chen K, Wang H. Recent Advances in Cell and Functional Biomaterial Treatment for Spinal Cord Injury. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5079153. [PMID: 35978649 PMCID: PMC9377911 DOI: 10.1155/2022/5079153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is a devastating central nervous system disease caused by accidental events, resulting in loss of sensory and motor function. Considering the multiple effects of primary and secondary injuries after spinal cord injury, including oxidative stress, tissue apoptosis, inflammatory response, and neuronal autophagy, it is crucial to understand the underlying pathophysiological mechanisms, local microenvironment changes, and neural tissue functional recovery for preparing novel treatment strategies. Treatment based on cell transplantation has become the forefront of spinal cord injury therapy. The transplanted cells provide physical and nutritional support for the damaged tissue. At the same time, the implantation of biomaterials with specific biological functions at the site of the SCI has also been proved to improve the local inhibitory microenvironment and promote axonal regeneration, etc. The combined transplantation of cells and functional biomaterials for SCI treatment can result in greater neuroprotective and regenerative effects by regulating cell differentiation, enhancing cell survival, and providing physical and directional support for axon regeneration and neural circuit remodeling. This article reviews the pathophysiology of the spinal cord, changes in the microenvironment after injury, and the mechanisms and strategies for spinal cord regeneration and repair. The article will focus on summarizing and discussing the latest intervention models based on cell and functional biomaterial transplantation and the latest progress in combinational therapies in SCI repair. Finally, we propose the future prospects and challenges of current treatment regimens for SCI repair, to provide references for scientists and clinicians to seek better SCI repair strategies in the future.
Collapse
Affiliation(s)
- Tianyi Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Wenhao Zhu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Xiaoyu Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Chuan He
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Xiaolong Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Qiang Xin
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Kexin Chen
- Institute of Translational Medicine, First Hospital of Jilin University, Changchun 130021, China
| | - Haifeng Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
5
|
Petrus-Reurer S, Romano M, Howlett S, Jones JL, Lombardi G, Saeb-Parsy K. Immunological considerations and challenges for regenerative cellular therapies. Commun Biol 2021; 4:798. [PMID: 34172826 PMCID: PMC8233383 DOI: 10.1038/s42003-021-02237-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
The central goal of regenerative medicine is to replace damaged or diseased tissue with cells that integrate and function optimally. The capacity of pluripotent stem cells to produce unlimited numbers of differentiated cells is of considerable therapeutic interest, with several clinical trials underway. However, the host immune response represents an important barrier to clinical translation. Here we describe the role of the host innate and adaptive immune responses as triggers of allogeneic graft rejection. We discuss how the immune response is determined by the cellular therapy. Additionally, we describe the range of available in vitro and in vivo experimental approaches to examine the immunogenicity of cellular therapies, and finally we review potential strategies to ameliorate immune rejection. In conclusion, we advocate establishment of platforms that bring together the multidisciplinary expertise and infrastructure necessary to comprehensively investigate the immunogenicity of cellular therapies to ensure their clinical safety and efficacy.
Collapse
Affiliation(s)
- Sandra Petrus-Reurer
- Department of Surgery, University of Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom.
| | - Marco Romano
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Sarah Howlett
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Joanne Louise Jones
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, and NIHR Cambridge Biomedical Research Centre, Cambridge, United Kingdom.
| |
Collapse
|
6
|
[Protective effect of transplantation of human oligodendrocyte precursor cells in a rat model of white matter injury]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23. [PMID: 33840415 PMCID: PMC8050556 DOI: 10.7499/j.issn.1008-8830.2011110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To study the effect of human oligodendrocyte precursor cell (hOPC) transplantation in the treatment of white matter injury (WMI). METHODS Neonatal rats were randomly divided into a sham-operation group, a model group, and a transplantation group (n=10 each). At the age of 3 days, the rats in the model group and the transplantation group were treated with right common carotid artery ligation, followed by hypoxia for 2 hours, to prepare a rat model of WMI. hOPCs were isolated from a spontaneously aborted human fetal brain at week 11 of gestation, and then hOPCs were cultured and transplanted into the rats with WMI. At 3 months after transplantation, the water maze test was performed to evaluate neurological function, and an electron microscope was used to observe myelin sheath thickness and proliferation. RESULTS The place navigation test using the Morris water maze showed that the model group had a significantly longer escape latency than the sham-operation group, and compared with the model group, the transplantation group had a significant reduction in escape latency (P < 0.05). To a certain degree, hOPC transplantation alleviated cognitive impairment in rats with WMI at the age of 90 days. The electron microscope images showed that hOPC transplantation promoted remyelination in the brain of WMI rats. Compared with the sham-operation group, the model group had a significant increase in the g-ratio (total axon diameter/total fiber diameter). Compared with the model group, the transplantation group had a significant reduction in the g-ratio (P < 0.05). CONCLUSIONS Intrathecal hOPC transplantation may alleviate neurological injury and promote remyelination in a rat model of WMI.
Collapse
|
7
|
Bell LA, Wallis GJ, Wilcox KS. Reactivity and increased proliferation of NG2 cells following central nervous system infection with Theiler's murine encephalomyelitis virus. J Neuroinflammation 2020; 17:369. [PMID: 33272299 PMCID: PMC7713670 DOI: 10.1186/s12974-020-02043-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/24/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Neuron-glial antigen 2 (NG2) cells are a glial cell type tiled throughout the gray and white matter of the central nervous system (CNS). NG2 cells are known for their ability to differentiate into oligodendrocytes and are commonly referred to as oligodendrocyte precursor cells. However, recent investigations have begun to identify additional functions of NG2 cells in CNS health and pathology. NG2 cells form physical and functional connections with neurons and other glial cell types throughout the CNS, allowing them to monitor and respond to the neural environment. Growing evidence indicates that NG2 cells become reactive under pathological conditions, though their specific roles are only beginning to be elucidated. While reactive microglia and astrocytes are well-established contributors to neuroinflammation and the development of epilepsy following CNS infection, the dynamics of NG2 cells remain unclear. Therefore, we investigated NG2 cell reactivity in a viral-induced mouse model of temporal lobe epilepsy. METHODS C57BL6/J mice were injected intracortically with Theiler's murine encephalomyelitis virus (TMEV) or PBS. Mice were graded twice daily for seizures between 3 and 7 days post-injection (dpi). At 4 and 14 dpi, brains were fixed and stained for NG2, the microglia/macrophage marker IBA1, and the proliferation marker Ki-67. Confocal z stacks were acquired in both the hippocampus and the overlying cortex. Total field areas stained by each cell marker and total field area of colocalized pixels between NG2 and Ki67 were compared between groups. RESULTS Both NG2 cells and microglia/macrophages displayed increased immunoreactivity and reactive morphologies in the hippocampus of TMEV-injected mice. While increased immunoreactivity for IBA1 was also present in the cortex, there was no significant change in NG2 immunoreactivity in the cortex following TMEV infection. Colocalization analysis for NG2 and Ki-67 revealed a significant increase in overlap between NG2 and Ki-67 in the hippocampus of TMEV-injected mice at both time points, but no significant differences in cortex. CONCLUSIONS NG2 cells acquire a reactive phenotype and proliferate in response to TMEV infection. These results suggest that NG2 cells alter their function in response to viral encephalopathy, making them potential targets to prevent the development of epilepsy following viral infection.
Collapse
Affiliation(s)
- Laura A Bell
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, 84112, USA
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, 84112, USA
| | - Glenna J Wallis
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, 84112, USA
| | - Karen S Wilcox
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT, 84112, USA.
- Interdepartmental Program in Neuroscience, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
8
|
Mozafari S, Baron-Van Evercooren A. Human stem cell-derived oligodendrocytes: From humanized animal models to cell therapy in myelin diseases. Semin Cell Dev Biol 2020; 116:53-61. [PMID: 33082116 DOI: 10.1016/j.semcdb.2020.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes are main targets in demyelinating and dysmyelinating diseases of the central nervous system (CNS), but are also involved in accidental, neurodegenerative and psychiatric disorders. The underlying pathology of these diseases is not fully understood and treatments are still lacking. The recent discovery of the induced pluripotent stem cell (iPSC) technology has open the possibility to address the biology of human oligodendroglial cells both in the dish and in vivo via engraftment in animal models, and paves the way for the development of treatment for myelin disorders. In this review, we make a short overview of the different sources human oligodendroglial cells, and animal models available for pre-clinical cell therapy. We discuss the anatomical and functional benefit of grafted iPSC-progenitors over their brain counterparts, their use in disease modeling and the missing gaps that still prevent to study their biology in the most integrated way, and to translate iPSC-stem cell based therapy to the clinic.
Collapse
Affiliation(s)
- Sabah Mozafari
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France
| | - Anne Baron-Van Evercooren
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France.
| |
Collapse
|
9
|
Zhang SZ, Wang QQ, Yang QQ, Gu HY, Yin YQ, Li YD, Hou JC, Chen R, Sun QQ, Sun YF, Hu G, Zhou JW. NG2 glia regulate brain innate immunity via TGF-β2/TGFBR2 axis. BMC Med 2019; 17:204. [PMID: 31727112 PMCID: PMC6857135 DOI: 10.1186/s12916-019-1439-x] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/01/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Brain innate immunity is vital for maintaining normal brain functions. Immune homeostatic imbalances play pivotal roles in the pathogenesis of neurological diseases including Parkinson's disease (PD). However, the molecular and cellular mechanisms underlying the regulation of brain innate immunity and their significance in PD pathogenesis are still largely unknown. METHODS Cre-inducible diphtheria toxin receptor (iDTR) and diphtheria toxin-mediated cell ablation was performed to investigate the impact of neuron-glial antigen 2 (NG2) glia on the brain innate immunity. RNA sequencing analysis was carried out to identify differentially expressed genes in mouse brain with ablated NG2 glia and lipopolysaccharide (LPS) challenge. Neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated mice were used to evaluate neuroinflammatory response in the presence or absence of NG2 glia. The survival of dopaminergic neurons or glial cell activation was evaluated by immunohistochemistry. Co-cultures of NG2 glia and microglia were used to examine the influence of NG2 glia to microglial activation. RESULTS We show that NG2 glia are required for the maintenance of immune homeostasis in the brain via transforming growth factor-β2 (TGF-β2)-TGF-β type II receptor (TGFBR2)-CX3C chemokine receptor 1 (CX3CR1) signaling, which suppresses the activation of microglia. We demonstrate that mice with ablated NG2 glia display a profound downregulation of the expression of microglia-specific signature genes and remarkable inflammatory response in the brain following exposure to endotoxin lipopolysaccharides. Gain- or loss-of-function studies show that NG2 glia-derived TGF-β2 and its receptor TGFBR2 in microglia are key regulators of the CX3CR1-modulated immune response. Furthermore, deficiency of NG2 glia contributes to neuroinflammation and nigral dopaminergic neuron loss in MPTP-induced mouse PD model. CONCLUSIONS These findings suggest that NG2 glia play a critical role in modulation of neuroinflammation and provide a compelling rationale for the development of new therapeutics for neurological disorders.
Collapse
Affiliation(s)
- Shu-Zhen Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Qin-Qin Wang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China.,Neurobiology Key Laboratory, Jining Medical University, Jining, 272067, Shandong, China
| | - Qiao-Qiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Huan-Yu Gu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yan-Qing Yin
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yan-Dong Li
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Jin-Can Hou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Rong Chen
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing-Qing Sun
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying-Feng Sun
- Center for Brain Disorders Research, Center of Parkinson's Disease, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100053, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jia-Wei Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China. .,School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China. .,Co-innovation Center of Neuroregeneration, School of Medicine, Nantong University, Nantong, 226001, Jiangsu, China. .,Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, 201210, China.
| |
Collapse
|
10
|
Pereira IM, Marote A, Salgado AJ, Silva NA. Filling the Gap: Neural Stem Cells as A Promising Therapy for Spinal Cord Injury. Pharmaceuticals (Basel) 2019; 12:ph12020065. [PMID: 31035689 PMCID: PMC6631328 DOI: 10.3390/ph12020065] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/15/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) can lead to severe motor, sensory and social impairments having a huge impact on patients’ lives. The complex and time-dependent SCI pathophysiology has been hampering the development of novel and effective therapies. Current treatment options include surgical interventions, to stabilize and decompress the spinal cord, and rehabilitative care, without providing a cure for these patients. Novel therapies have been developed targeting different stages during trauma. Among them, cell-based therapies hold great potential for tissue regeneration after injury. Neural stem cells (NSCs), which are multipotent cells with inherent differentiation capabilities committed to the neuronal lineage, are especially relevant to promote and reestablish the damaged neuronal spinal tracts. Several studies demonstrate the regenerative effects of NSCs in SCI after transplantation by providing neurotrophic support and restoring synaptic connectivity. Therefore, human clinical trials have already been launched to assess safety in SCI patients. Here, we review NSC-based experimental studies in a SCI context and how are they currently being translated into human clinical trials.
Collapse
Affiliation(s)
- Inês M Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Ana Marote
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
11
|
Dhar C, Sasmal A, Varki A. From "Serum Sickness" to "Xenosialitis": Past, Present, and Future Significance of the Non-human Sialic Acid Neu5Gc. Front Immunol 2019; 10:807. [PMID: 31057542 PMCID: PMC6481270 DOI: 10.3389/fimmu.2019.00807] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/26/2019] [Indexed: 01/01/2023] Open
Abstract
The description of "serum sickness" more than a century ago in humans transfused with animal sera eventually led to identification of a class of human antibodies directed against glycans terminating in the common mammalian sialic acid N-Glycolylneuraminic acid (Neu5Gc), hereafter called "Neu5Gc-glycans." The detection of such glycans in malignant and fetal human tissues initially raised the possibility that it was an oncofetal antigen. However, "serum sickness" antibodies were also noted in various human disease states. These findings spurred further research on Neu5Gc, and the discovery that it is not synthesized in the human body due to a human-lineage specific genetic mutation in the enzyme CMAH. However, with more sensitive techniques Neu5Gc-glycans were detected in smaller quantities on certain human cell types, particularly epithelia and endothelia. The likely explanation is metabolic incorporation of Neu5Gc from dietary sources, especially red meat of mammalian origin. This incorporated Neu5Gc on glycans appears to be the first example of a "xeno-autoantigen," against which varying levels of "xeno-autoantibodies" are present in all humans. The resulting chronic inflammation or "xenosialitis" may have important implications in human health and disease, especially in conditions known to be aggravated by consumption of red meat. In this review, we will cover the early history of the discovery of "serum sickness" antibodies, the subsequent recognition that they were partly directed against Neu5Gc-glycans, the discovery of the genetic defect eliminating Neu5Gc production in humans, and the later recognition that this was not an oncofetal antigen but the first example of a "xeno-autoantigen." Further, we will present comments about implications for disease risks associated with red meat consumption such as cancer and atherosclerosis. We will also mention the potential utility of these anti-Neu5Gc-glycan antibodies in cancer immunotherapy and provide some suggestions and perspectives for the future. Other reviews in this special issue cover many other aspects of this unusual pathological process, for which there appears to be no other described precedent.
Collapse
Affiliation(s)
- Chirag Dhar
- Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, United States
| | - Aniruddha Sasmal
- Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, United States
| | - Ajit Varki
- Departments of Medicine and Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
12
|
Scheld M, Fragoulis A, Nyamoya S, Zendedel A, Denecke B, Krauspe B, Teske N, Kipp M, Beyer C, Clarner T. Mitochondrial Impairment in Oligodendroglial Cells Induces Cytokine Expression and Signaling. J Mol Neurosci 2018; 67:265-275. [PMID: 30547416 DOI: 10.1007/s12031-018-1236-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/28/2018] [Indexed: 12/14/2022]
Abstract
Widespread inflammatory lesions within the central nervous system grey and white matter are major hallmarks of multiple sclerosis. The development of full-blown demyelinating multiple sclerosis lesions might be preceded by preactive lesions which are characterized by focal microglia activation in close spatial relation to apoptotic oligodendrocytes. In this study, we investigated the expression of signaling molecules of oligodendrocytes that might be involved in initial microglia activation during preactive lesion formation. Sodium azide was used to trigger mitochondrial impairment and cellular stress in oligodendroglial cells in vitro. Among various chemokines and cytokines, IL6 was identified as a possible oligodendroglial cell-derived signaling molecule in response to cellular stress. Relevance of this finding for lesion development was further explored in the cuprizone model by applying short-term cuprizone feeding (2-4 days) on male C57BL/6 mice and subsequent analysis of gene expression, in situ hybridization and histology. Additionally, we analyzed the possible signaling of stressed oligodendroglial cells in vitro as well as in the cuprizone mouse model. In vitro, conditioned medium of stressed oligodendroglial cells triggered the activation of microglia cells. In cuprizone-fed animals, IL6 expression in oligodendrocytes was found in close vicinity of activated microglia cells. Taken together, our data support the view that stressed oligodendrocytes have the potential to activate microglia cells through a specific cocktail of chemokines and cytokines among IL6. Further studies will have to identify the temporal activation pattern of these signaling molecules, their cellular sources, and impact on neuroinflammation.
Collapse
Affiliation(s)
- Miriam Scheld
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Athanassios Fragoulis
- Department of Anatomy and Cell Biology, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Stella Nyamoya
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.,Department of Neuroanatomy, Faculty of Medicine, Ludwig-Maximilians-University of Munich, 80336, Munich, Germany
| | - Adib Zendedel
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Bernd Denecke
- IZKF Genomics Facility, Interdisciplinary Center for Clinical Research, RWTH Aachen University, 52074, Aachen, Germany
| | - Barbara Krauspe
- Clinic for Gynaecology and Obstetrics, Faculty of Medicine, RWTH Aachen University, 52074, Aachen, Germany
| | - Nico Teske
- Department of Neuroanatomy, Faculty of Medicine, Ludwig-Maximilians-University of Munich, 80336, Munich, Germany
| | - Markus Kipp
- Institute of Anatomy, Faculty of Medicine, University of Rostock, 18057, Rostock, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Tim Clarner
- Institute of Neuroanatomy, Faculty of Medicine, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| |
Collapse
|
13
|
Ahuja CS, Nori S, Tetreault L, Wilson J, Kwon B, Harrop J, Choi D, Fehlings MG. Traumatic Spinal Cord Injury-Repair and Regeneration. Neurosurgery 2017; 80:S9-S22. [PMID: 28350947 DOI: 10.1093/neuros/nyw080] [Citation(s) in RCA: 566] [Impact Index Per Article: 70.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/12/2017] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Traumatic spinal cord injuries (SCI) have devastating consequences for the physical, financial, and psychosocial well-being of patients and their caregivers. Expediently delivering interventions during the early postinjury period can have a tremendous impact on long-term functional recovery. PATHOPHYSIOLOGY This is largely due to the unique pathophysiology of SCI where the initial traumatic insult (primary injury) is followed by a progressive secondary injury cascade characterized by ischemia, proapoptotic signaling, and peripheral inflammatory cell infiltration. Over the subsequent hours, release of proinflammatory cytokines and cytotoxic debris (DNA, ATP, reactive oxygen species) cyclically adds to the harsh postinjury microenvironment. As the lesions mature into the chronic phase, regeneration is severely impeded by the development of an astroglial-fibrous scar surrounding coalesced cystic cavities. Addressing these challenges forms the basis of current and upcoming treatments for SCI. MANAGEMENT This paper discusses the evidence-based management of a patient with SCI while emphasizing the importance of early definitive care. Key neuroprotective therapies are summarized including surgical decompression, methylprednisolone, and blood pressure augmentation. We then review exciting neuroprotective interventions on the cusp of translation such as Riluzole, Minocycline, magnesium, therapeutic hypothermia, and CSF drainage. We also explore the most promising neuroregenerative strategies in trial today including Cethrin™, anti-NOGO antibody, cell-based approaches, and bioengineered biomaterials. Each section provides a working knowledge of the key preclinical and patient trials relevant to clinicians while highlighting the pathophysiologic rationale for the therapies. CONCLUSION We conclude with our perspectives on the future of treatment and research in this rapidly evolving field.
Collapse
Affiliation(s)
- Christopher S Ahuja
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada.,Department of Genetics and Development, University of Toronto, Toronto, Canada
| | - Satoshi Nori
- Department of Genetics and Development, University of Toronto, Toronto, Canada
| | | | - Jefferson Wilson
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada.,Spine Program, University of Toronto, Toronto, Canada
| | - Brian Kwon
- Vancouver Spine Institute, Vancouver General Hospital, Vancouver, Canada.,Department of Surgery, University of British Columbia, Vancouver, Canada
| | - James Harrop
- Thomas Jefferson University Hospital, Philadelphia, Pennsylvania
| | - David Choi
- National Hospital for Neurology and Neurosurgery, University College London, London, England
| | - Michael G Fehlings
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Surgery, University of Toronto, Toronto, Canada.,Spine Program, University of Toronto, Toronto, Canada.,Department of Genetics and Development, University of Toronto, Toronto, Canada
| |
Collapse
|
14
|
Martínez-Cerdeño V, Barrilleaux BL, McDonough A, Ariza J, Yuen BTK, Somanath P, Le CT, Steward C, Horton-Sparks K, Knoepfler PS. Behavior of Xeno-Transplanted Undifferentiated Human Induced Pluripotent Stem Cells Is Impacted by Microenvironment Without Evidence of Tumors. Stem Cells Dev 2017; 26:1409-1423. [PMID: 28693365 DOI: 10.1089/scd.2017.0059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human pluripotent stem cells (hPSC) have great clinical potential through the use of their differentiated progeny, a population in which there is some concern over risks of tumorigenicity or other unwanted cellular behavior due to residual hPSC. Preclinical studies using human stem cells are most often performed within a xenotransplant context. In this study, we sought to measure how undifferentiated hPSC behave following xenotransplant. We directly transplanted undifferentiated human induced pluripotent stem cells (hIPSC) and human embryonic stem cells (hESC) into the adult mouse brain ventricle and analyzed their fates. No tumors or precancerous lesions were present at more than one year after transplantation. This result differed with the tumorigenic capacity we observed after allotransplantation of mouse ESC into the mouse brain. A substantial population of cellular derivatives of undifferentiated hESC and hIPSC engrafted, survived, and migrated within the mouse brain parenchyma. Within brain structures, transplanted cell distribution followed a very specific pattern, suggesting the existence of distinct microenvironments that offer different degrees of permissibility for engraftment. Most of the transplanted hESC and hIPSC that developed into brain cells were NeuN+ neuronal cells, and no astrocytes were detected. Substantial cell and nuclear fusion occurred between host and transplanted cells, a phenomenon influenced by microenvironment. Overall, hIPSC appear to be largely functionally equivalent to hESC in vivo. Altogether, these data bring new insights into the behavior of stem cells without prior differentiation following xenotransplantation into the adult brain.
Collapse
Affiliation(s)
- Veronica Martínez-Cerdeño
- 1 Department of Pathology and Laboratory Medicine, University of California Davis School of Medicine , Sacramento, California.,2 Institute for Regenerative Cures, University of California Davis School of Medicine , Sacramento, California.,3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California
| | - Bonnie L Barrilleaux
- 2 Institute for Regenerative Cures, University of California Davis School of Medicine , Sacramento, California.,3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California.,4 Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine , Sacramento, California
| | - Ashley McDonough
- 3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California
| | - Jeanelle Ariza
- 3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California
| | - Benjamin T K Yuen
- 2 Institute for Regenerative Cures, University of California Davis School of Medicine , Sacramento, California.,3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California.,4 Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine , Sacramento, California
| | - Priyanka Somanath
- 2 Institute for Regenerative Cures, University of California Davis School of Medicine , Sacramento, California.,3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California.,4 Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine , Sacramento, California
| | - Catherine T Le
- 2 Institute for Regenerative Cures, University of California Davis School of Medicine , Sacramento, California.,3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California.,4 Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine , Sacramento, California
| | - Craig Steward
- 3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California
| | - Kayla Horton-Sparks
- 3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California
| | - Paul S Knoepfler
- 2 Institute for Regenerative Cures, University of California Davis School of Medicine , Sacramento, California.,3 Institute of Pediatric Regenerative Medicine , Shriners Hospital for Children, Northern California, Sacramento, California.,4 Department of Cell Biology and Human Anatomy, University of California Davis School of Medicine , Sacramento, California
| |
Collapse
|
15
|
Abstract
Traumatic spinal cord injuries (SCIs) affect 1.3 million North Americans, producing devastating physical, social, and vocational impairment. Pathophysiologically, the initial mechanical trauma is followed by a significant secondary injury which includes local ischemia, pro-apoptotic signaling, release of cytotoxic factors, and inflammatory cell infiltration. Expedient delivery of medical and surgical care during this critical period can improve long-term functional outcomes, engendering the concept of "Time is Spine". We emphasize the importance of expeditious care while outlining the initial clinical and radiographic assessment of patients. Key evidence-based early interventions (surgical decompression, blood pressure augmentation, and methylprednisolone) are also reviewed, including findings of the landmark Surgical Timing in Acute Spinal Cord Injury Study (STASCIS). We then describe other neuroprotective approaches on the edge of translation such as the sodium-channel blocker riluzole, the anti-inflammatory minocycline, and therapeutic hypothermia. We also review promising neuroregenerative therapies that are likely to influence management practices over the next decade including chondroitinase, Rho-ROCK pathway inhibition, and bioengineered strategies. The importance of emerging neural stem cell therapies to remyelinate denuded axons and regenerate neural circuits is also discussed. Finally, we outline future directions for research and patient care.
Collapse
Affiliation(s)
- Christopher S Ahuja
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Allan R Martin
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Michael Fehlings
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada; McEwen Centre for Regenerative Medicine, UHN, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, University of Toronto, Toronto, Ontario, Canada; Spine Program, University of Toronto, Toronto, Ontario, Canada; McLaughlin Center in Molecular Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
16
|
Ahuja CS, Fehlings M. Concise Review: Bridging the Gap: Novel Neuroregenerative and Neuroprotective Strategies in Spinal Cord Injury. Stem Cells Transl Med 2016; 5:914-24. [PMID: 27130222 DOI: 10.5966/sctm.2015-0381] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/07/2016] [Indexed: 12/13/2022] Open
Abstract
UNLABELLED Spinal cord injuries (SCIs) result in devastating lifelong disability for patients and their families. The initial mechanical trauma is followed by a damaging secondary injury cascade involving proapoptotic signaling, ischemia, and inflammatory cell infiltration. Ongoing cellular necrosis releases ATP, DNA, glutamate, and free radicals to create a cytotoxic postinjury milieu. Long-term regeneration of lost or injured networks is further impeded by cystic cavitation and the formation of an inhibitory glial-chondroitin sulfate proteoglycan scar. In this article, we discuss important neuroprotective interventions currently applied in clinical practice, including surgical decompression, blood pressure augmentation, and i.v. methylprednisolone. We then explore exciting translational therapies on the horizon, such as riluzole, minocycline, fibroblast growth factor, magnesium, and hypothermia. Finally, we summarize the key neuroregenerative strategies of the next decade, including glial scar degradation, Rho-ROCK inhibition, cell-based therapies, and novel bioengineered adjuncts. Throughout, we emphasize the need for combinatorial approaches to this multifactorial problem and discuss relevant studies at the forefront of translation. We conclude by providing our perspectives on the future direction of SCI research. SIGNIFICANCE Spinal cord injuries (SCIs) result in devastating, lifelong disability for patients and their families. This article discusses important neuroprotective interventions currently applied in clinical practice, including surgical decompression, blood pressure augmentation, and i.v. methylprednisolone. Translational therapies on the horizon are discussed, such as riluzole, minocycline, fibroblast growth factor, magnesium, and hypothermia. The key neuroregenerative strategies of the next decade are summarized, including glial scar degradation, Rho-ROCK inhibition, cell-based therapies, and novel bioengineered adjuncts. The need for combinatorial approaches to this multifactorial problem is emphasized, relevant studies at the forefront of translation are discussed, and perspectives on the future direction of SCI research are presented.
Collapse
Affiliation(s)
- Christopher S Ahuja
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Michael Fehlings
- Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Ontario, Canada Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada McEwen Centre for Regenerative Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada Department of Surgery, University of Toronto, Toronto, Ontario, Canada Spine Program, University of Toronto, Toronto, Ontario, Canada McLaughlin Centre for Molecular Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
17
|
|
18
|
Lipopolysaccharide Upregulates the Expression of CINC-3 and LIX in Primary NG2 Cells. Neurochem Res 2016; 41:1448-57. [PMID: 26842931 DOI: 10.1007/s11064-016-1856-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/01/2016] [Accepted: 01/28/2016] [Indexed: 10/22/2022]
Abstract
Numerous NG2 cells, also called oligodendrocyte progenitor cells (OPCs), exist ubiquitously in the gray and white matter in the adult central nervous system (CNS). Although NG2 cells could become active by upregulation of NG2 expression and hypertrophy or extension of their processes under various neuropathological conditions, their actual role in the brain remains to be illustrated. In view of the fact that the synergy of cytokine and chemokine networks plays an important role in CNS inflammation and immunity, we have assumed that the NG2 cells might take part in brain inflammation and immunity by making a contribution to the pool of cytokines or chemokines. In the current study, NG2-expressing OPCs were prepared from cerebral hemispheres of postnatal day 0 or 1 Sprague-Dawley rats. Our results showed that NG2-expressing OPCs, verified by immunohistological staining of anti-NG2 antibody and anti-platelet-derived growth factor receptor alpha (PDGFRα) antibody, presented binding affinity to lipopolysaccharide (LPS), a commonly used stimulator in a neuroinflammatory model. Using cytokine antibody array, QPCR and ELISA, we have further shown that LPS could upregulate the expression of cytokine induced neutrophil chemoattractant-3 (CINC-3) and LPS induced CXC chemokine (LIX) in primary NG2-expressing OPCs, without the alteration in cell number of NG2-expressing OPCs. In addition, the cells bearing the receptor for these two cytokines included microglia and OPCs. Taken together, our results suggest that NG2-expressing OPCs could response to LPS and may take part in neuroinflammatory process, through secreting cytokines and chemokines to exert an effect on target cells (OPCs and microglia).
Collapse
|
19
|
Haworth R, Sharpe M. The issue of immunology in stem cell therapies: a pharmaceutical perspective. Regen Med 2016; 10:231-4. [PMID: 25933230 DOI: 10.2217/rme.14.50] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cautious approaches in the clinic are currently proposed, supported by relevant in vitro, in vivo and published data. Key to developing our understanding of the risks of immune rejection of stem cell based therapies will be the inclusion of immunological endpoints in clinical trials and the sharing of data. There is likely not a one-size-fits all strategy but one dependent on cell therapy, mode of action and disease indication.
Collapse
Affiliation(s)
- Richard Haworth
- Safety Assessment, GlaxoSmithKline R&D, Park Road, Ware, Herts., SG12 0DP, UK
| | | |
Collapse
|
20
|
Zeis T, Enz L, Schaeren-Wiemers N. The immunomodulatory oligodendrocyte. Brain Res 2015; 1641:139-148. [PMID: 26423932 DOI: 10.1016/j.brainres.2015.09.021] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 09/17/2015] [Accepted: 09/20/2015] [Indexed: 01/12/2023]
Abstract
Oligodendrocytes, the myelinating glial cells of the central nervous system (CNS), are due to their high specialization and metabolic needs highly vulnerable to various insults. This led to a general view that oligodendrocytes are defenseless victims during brain damage such as occurs in acute and chronic CNS inflammation. However, this view is challenged by increasing evidence that oligodendrocytes are capable of expressing a wide range of immunomodulatory molecules. They express various cytokines and chemokines (e.g. Il-1β, Il17A, CCL2, CXCL10), antigen presenting molecules (MHC class I and II) and co-stimulatory molecules (e.g. CD9, CD81), complement and complement receptor molecules (e.g. C1s, C2 and C3, C1R), complement regulatory molecules (e.g. CD46, CD55, CD59), tetraspanins (e.g. TSPAN2), neuroimmune regulatory proteins (e.g. CD200, CD47) as well as extracellular matrix proteins (e.g. VCAN) and many others. Their potential immunomodulatory properties can, at specific times and locations, influence ongoing immune processes as shown by numerous publications. Therefore, oligodendrocytes are well capable of immunomodulation, especially during the initiation or resolution of immune processes in which subtle signaling might tip the scale. A better understanding of the immunomodulatory oligodendrocyte can help to invent new, innovative therapeutic interventions in various diseases such as Multiple Sclerosis. This article is part of a Special Issue entitled SI: Myelin Evolution.
Collapse
Affiliation(s)
- Thomas Zeis
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Lukas Enz
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nicole Schaeren-Wiemers
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| |
Collapse
|
21
|
Stem cell based therapies for age-related macular degeneration: The promises and the challenges. Prog Retin Eye Res 2015; 48:1-39. [PMID: 26113213 DOI: 10.1016/j.preteyeres.2015.06.004] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 12/21/2022]
|
22
|
Buzhor E, Leshansky L, Blumenthal J, Barash H, Warshawsky D, Mazor Y, Shtrichman R. Cell-based therapy approaches: the hope for incurable diseases. Regen Med 2015; 9:649-72. [PMID: 25372080 DOI: 10.2217/rme.14.35] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cell therapies aim to repair the mechanisms underlying disease initiation and progression, achieved through trophic effect or by cell replacement. Multiple cell types can be utilized in such therapies, including stem, progenitor or primary cells. This review covers the current state of cell therapies designed for the prominent disorders, including cardiovascular, neurological (Parkinson's disease, amyotrophic lateral sclerosis, stroke, spinal cord injury), autoimmune (Type 1 diabetes, multiple sclerosis, Crohn's disease), ophthalmologic, renal, liver and skeletal (osteoarthritis) diseases. Various cell therapies have reached advanced clinical trial phases with potential marketing approvals in the near future, many of which are based on mesenchymal stem cells. Advances in pluripotent stem cell research hold great promise for regenerative medicine. The information presented in this review is based on the analysis of the cell therapy collection detailed in LifeMap Discovery(®) (LifeMap Sciences Inc., USA) the database of embryonic development, stem cell research and regenerative medicine.
Collapse
|
23
|
Casarosa S, Bozzi Y, Conti L. Neural stem cells: ready for therapeutic applications? MOLECULAR AND CELLULAR THERAPIES 2014; 2:31. [PMID: 26056597 PMCID: PMC4452059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 10/05/2014] [Indexed: 11/21/2023]
Abstract
Neural stem cells (NSCs) offer a unique and powerful tool for basic research and regenerative medicine. However, the challenges that scientists face in the comprehension of the biology and physiological function of these cells are still many. Deciphering NSCs fundamental biological aspects represents indeed a crucial step to control NSCs fate and functional integration following transplantation, and is essential for a safe and appropriate use of NSCs in injury/disease conditions. In this review, we focus on the biological properties of NSCs and discuss how these cells may be exploited to provide effective therapies for neurological disorders. We also review and discuss ongoing NSC-based clinical trials for these diseases.
Collapse
Affiliation(s)
- Simona Casarosa
- Center for Integrative Biology, Università degli Studi di Trento, Via Sommarive 9, Povo-Trento, 38123 Italy
| | - Yuri Bozzi
- Center for Integrative Biology, Università degli Studi di Trento, Via Sommarive 9, Povo-Trento, 38123 Italy
| | - Luciano Conti
- Center for Integrative Biology, Università degli Studi di Trento, Via Sommarive 9, Povo-Trento, 38123 Italy
| |
Collapse
|
24
|
Casarosa S, Bozzi Y, Conti L. Neural stem cells: ready for therapeutic applications? MOLECULAR AND CELLULAR THERAPIES 2014; 2:31. [PMID: 26056597 PMCID: PMC4452059 DOI: 10.1186/2052-8426-2-31] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 10/05/2014] [Indexed: 12/14/2022]
Abstract
Neural stem cells (NSCs) offer a unique and powerful tool for basic research and regenerative medicine. However, the challenges that scientists face in the comprehension of the biology and physiological function of these cells are still many. Deciphering NSCs fundamental biological aspects represents indeed a crucial step to control NSCs fate and functional integration following transplantation, and is essential for a safe and appropriate use of NSCs in injury/disease conditions. In this review, we focus on the biological properties of NSCs and discuss how these cells may be exploited to provide effective therapies for neurological disorders. We also review and discuss ongoing NSC-based clinical trials for these diseases.
Collapse
Affiliation(s)
- Simona Casarosa
- Center for Integrative Biology, Università degli Studi di Trento, Via Sommarive 9, Povo-Trento, 38123 Italy
| | - Yuri Bozzi
- Center for Integrative Biology, Università degli Studi di Trento, Via Sommarive 9, Povo-Trento, 38123 Italy
| | - Luciano Conti
- Center for Integrative Biology, Università degli Studi di Trento, Via Sommarive 9, Povo-Trento, 38123 Italy
| |
Collapse
|
25
|
Tan Y, Ooi S, Wang L. Immunogenicity and tumorigenicity of pluripotent stem cells and their derivatives: genetic and epigenetic perspectives. Curr Stem Cell Res Ther 2014; 9:63-72. [PMID: 24160683 PMCID: PMC3873036 DOI: 10.2174/1574888x113086660068] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Revised: 10/19/2013] [Accepted: 10/22/2013] [Indexed: 12/18/2022]
Abstract
One aim of stem cell-based therapy is to utilize pluripotent stem cells (PSCs) as a supplementary source of cells
to repair or replace tissues or organs that have ceased to function due to severe tissue damage. However, PSC-based therapy
requires extensive research to ascertain if PSC derivatives are functional without the risk of tumorigenicity, and also
do not engender severe immune rejection that threatens graft survival and function. Recently, the suitability of induced
pluripotent stem cells applied for patient-tailored cell therapy has been questioned since the discovery of several genetic
and epigenetic aberrations during the reprogramming process. Hence, it is crucial to understand the effect of these abnormalities
on the immunogenicity and survival of PSC grafts. As induced PSC-based therapy represents a hallmark for the
potential solution to prevent and arrest immune rejection, this review also summarizes several up-to-date key findings in
the field.
Collapse
Affiliation(s)
| | | | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Road, Ottawa K1H8M5, Canada.
| |
Collapse
|
26
|
Nguyen HX, Nekanti U, Haus DL, Funes G, Moreno D, Kamei N, Cummings BJ, Anderson AJ. Induction of early neural precursors and derivation of tripotent neural stem cells from human pluripotent stem cells under xeno-free conditions. J Comp Neurol 2014; 522:2767-83. [DOI: 10.1002/cne.23604] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 04/04/2014] [Accepted: 04/04/2014] [Indexed: 02/06/2023]
Affiliation(s)
- Hal X. Nguyen
- Physical Medicine & Rehabilitation; University of California; Irvine California
- Anatomy and Neurobiology; University of California; Irvine California
- Sue and Bill Gross Stem Cell Research Center; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine California
| | - Usha Nekanti
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine California
| | - Daniel L. Haus
- Anatomy and Neurobiology; University of California; Irvine California
- Sue and Bill Gross Stem Cell Research Center; University of California; Irvine California
| | - Gabrielle Funes
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine California
| | - Denisse Moreno
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine California
| | - Noriko Kamei
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine California
| | - Brian J. Cummings
- Physical Medicine & Rehabilitation; University of California; Irvine California
- Anatomy and Neurobiology; University of California; Irvine California
- Sue and Bill Gross Stem Cell Research Center; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine California
| | - Aileen J. Anderson
- Physical Medicine & Rehabilitation; University of California; Irvine California
- Anatomy and Neurobiology; University of California; Irvine California
- Sue and Bill Gross Stem Cell Research Center; University of California; Irvine California
- Institute for Memory Impairments and Neurological Disorders; University of California; Irvine California
| |
Collapse
|
27
|
Scheiner ZS, Talib S, Feigal EG. The potential for immunogenicity of autologous induced pluripotent stem cell-derived therapies. J Biol Chem 2013; 289:4571-7. [PMID: 24362036 DOI: 10.1074/jbc.r113.509588] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) technology offers the promise of immune-matched cell therapies for a wide range of diseases and injuries. It is generally assumed that cells derived from autologous iPSCs will be immune-privileged. However, there are reasons to question this assumption, including recent studies that have tested iPSC immunogenicity in various ways with conflicting results. Understanding the risk of an immune response and developing strategies to minimize it will be important steps before clinical testing. Here, we review the evidence for autologous iPSC immunogenicity, its potential causes, and approaches for assessment and mitigation.
Collapse
Affiliation(s)
- Zachary S Scheiner
- From the California Institute for Regenerative Medicine, San Francisco, California 94107
| | | | | |
Collapse
|
28
|
Alsanie WF, Niclis JC, Petratos S. Human embryonic stem cell-derived oligodendrocytes: protocols and perspectives. Stem Cells Dev 2013; 22:2459-76. [PMID: 23621561 PMCID: PMC3760471 DOI: 10.1089/scd.2012.0520] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 04/26/2013] [Indexed: 12/19/2022] Open
Abstract
Oligodendrocytes play a fundamental supportive role in the mammalian central nervous system (CNS) as the myelinating-glial cells. Disruption of fast axonal transport mechanisms can occur as a consequence of mature oligodendrocyte loss following spinal cord injury, stroke, or due to neuroinflammatory conditions, such as multiple sclerosis. As a result of the limited remyelination ability in the CNS after injury or disease, human embryonic stem cells (hESCs) may prove to be a promising option for the generation and replacement of mature oligodendrocytes. Moreover, hESC-derived oligodendrocytes may be experimentally utilized to unravel fundamental questions of oligodendrocyte development, along with their therapeutic potential through growth factor support of axons and neurons. However, an intensive characterization and examination of hESC-derived oligodendrocytes prior to preclinical or clinical trials is required to facilitate greater success in their integration following cellular replacement therapy (CRT). Currently, the protocols utilized to derive oligodendrocytes from hESCs consist of significant variations in culture style, time-length of differentiation, and the provision of growth factors in culture. Further, these differing protocols also report disparate patterns in the expression of oligodendroglial markers by these derived oligodendrocytes, throughout their differentiation in culture. We have comprehensively reviewed the published protocols describing the derivation of oligodendrocytes from hESCs and the studies that examine their efficacy to remyelinate, along with the fundamental issues of their safety as a viable CRT. Additionally, this review will highlight particular issues of concern and suggestions for troubleshooting to provide investigators critical information for the future improvement of establishing in vitro hESC-derived oligodendrocytes.
Collapse
Affiliation(s)
- Walaa F Alsanie
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia.
| | | | | |
Collapse
|
29
|
Abbasalizadeh S, Baharvand H. Technological progress and challenges towards cGMP manufacturing of human pluripotent stem cells based therapeutic products for allogeneic and autologous cell therapies. Biotechnol Adv 2013; 31:1600-23. [PMID: 23962714 DOI: 10.1016/j.biotechadv.2013.08.009] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 06/20/2013] [Accepted: 08/12/2013] [Indexed: 12/16/2022]
Abstract
Recent technological advances in the generation, characterization, and bioprocessing of human pluripotent stem cells (hPSCs) have created new hope for their use as a source for production of cell-based therapeutic products. To date, a few clinical trials that have used therapeutic cells derived from hESCs have been approved by the Food and Drug Administration (FDA), but numerous new hPSC-based cell therapy products are under various stages of development in cell therapy-specialized companies and their future market is estimated to be very promising. However, the multitude of critical challenges regarding different aspects of hPSC-based therapeutic product manufacturing and their therapies have made progress for the introduction of new products and clinical applications very slow. These challenges include scientific, technological, clinical, policy, and financial aspects. The technological aspects of manufacturing hPSC-based therapeutic products for allogeneic and autologous cell therapies according to good manufacturing practice (cGMP) quality requirements is one of the most important challenging and emerging topics in the development of new hPSCs for clinical use. In this review, we describe main critical challenges and highlight a series of technological advances in all aspects of hPSC-based therapeutic product manufacturing including clinical grade cell line development, large-scale banking, upstream processing, downstream processing, and quality assessment of final cell therapeutic products that have brought hPSCs closer to clinical application and commercial cGMP manufacturing.
Collapse
Affiliation(s)
- Saeed Abbasalizadeh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | |
Collapse
|
30
|
O'Connor MD. The 3R principle: advancing clinical application of human pluripotent stem cells. Stem Cell Res Ther 2013; 4:21. [PMID: 23510719 PMCID: PMC3706962 DOI: 10.1186/scrt169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The first derivation of human embryonic stem cells brought with it a clear understanding that animal models of human disease might be replaced by an unlimited supply of human cells for research, drug discovery, and drug development. With the advent of clinical trials using human pluripotent stem cell-based therapies, it is both timely and relevant to reflect on factors that will facilitate future translation of this technology. Human pluripotent cells are increasingly being used to investigate the molecular mechanisms that underpin normal and pathological human development. Their differentiated progeny are also being used to identify novel pharmaceuticals, to screen for toxic effects of known chemicals, and to investigate cell or tissue transplantation strategies. The intrinsic assumption of these research efforts is that the information gained from these studies will be more accurate, and therefore of greater relevance, than traditional investigations based on animal models of human disease and injury. This review will therefore evaluate how animals and animal-derived products are used for human pluripotent stem cell research, and will indicate how efforts to further reduce or remove animals and animal products from this research will increase the clinical translation of human pluripotent stem cell technologies through drug discovery, toxicology screening, and cell replacement therapies.
Collapse
|
31
|
Li J, Lepski G. Cell transplantation for spinal cord injury: a systematic review. BIOMED RESEARCH INTERNATIONAL 2013; 2013:786475. [PMID: 23484157 PMCID: PMC3581246 DOI: 10.1155/2013/786475] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 11/16/2012] [Accepted: 12/11/2012] [Indexed: 02/07/2023]
Abstract
Cell transplantation, as a therapeutic intervention for spinal cord injury (SCI), has been extensively studied by researchers in recent years. A number of different kinds of stem cells, neural progenitors, and glial cells have been tested in basic research, and most have been excluded from clinical studies because of a variety of reasons, including safety and efficacy. The signaling pathways, protein interactions, cellular behavior, and the differentiated fates of experimental cells have been studied in vitro in detail. Furthermore, the survival, proliferation, differentiation, and effects on promoting functional recovery of transplanted cells have also been examined in different animal SCI models. However, despite significant progress, a "bench to bedside" gap still exists. In this paper, we comprehensively cover publications in the field from the last years. The most commonly utilized cell lineages were covered in this paper and specific areas covered include survival of grafted cells, axonal regeneration and remyelination, sensory and motor functional recovery, and electrophysiological improvements. Finally we also review the literature on the in vivo tracking techniques for transplanted cells.
Collapse
Affiliation(s)
- Jun Li
- Department of Neurosurgery, Eberhard Karls University, 72076 Tübingen, Germany
- Department of Spine Surgery, The Affiliated Hospital of Luzhou Medical College, 646000 Luzhou, China
| | - Guilherme Lepski
- Department of Neurosurgery, Eberhard Karls University, 72076 Tübingen, Germany
- Division of Neurosurgery, Department of Neurology, Faculdade de Medicina, Universidade de São Paulo, Avnida Dr. Enéas de Carvalho Aguiar 255, 05403-000 São Paulo, SP, Brazil
| |
Collapse
|
32
|
Human neural stem/progenitor cells derived from embryonic stem cells and fetal nervous system present differences in immunogenicity and immunomodulatory potentials in vitro. Stem Cell Res 2013; 10:325-37. [PMID: 23416350 DOI: 10.1016/j.scr.2013.01.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 12/18/2012] [Accepted: 01/02/2013] [Indexed: 12/29/2022] Open
Abstract
To develop cell therapies for damaged nervous tissue with human neural stem/progenitor cells (hNPCs), the risk of an immune response and graft rejection must be considered. There are conflicting results and lack of knowledge concerning the immunocompetence of hNPCs of different origin. Here, we studied the immunogenicity and immunomodulatory potentials of hNPCs cultured under equivalent conditions after derivation from human embryonic stem cells (hESC-NPCs) or human fetal spinal cord tissue (hfNPCs). The expression patterns of human leukocyte antigen, co-stimulatory and adhesion molecules in hESC-NPCs and hfNPCs were relatively similar and mostly not affected by inflammatory cytokines. Unstimulated hfNPCs secreted more transforming growth factor-β1 (TGF-β1) and β2 but similar level of interleukin (IL)-10 compared to hESC-NPCs. In contrast to hfNPCs, hESC-NPCs displayed 4-6 fold increases in TGF-β1, TGF-β2 and IL-10 under inflammatory conditions. Both hNPCs reduced the alloreaction between allogeneic peripheral blood mononuclear cells (PBMCs) and up-regulated CD4(+)CD25(+)forkhead box P3 (FOXP3)(+) T cells. However, hESC-NPCs but not hfNPCs dose-dependently triggered PBMC proliferation, which at least partly may be due to TGF-β signaling. To conclude, hESC-NPCs and hfNPCs displayed similarities but also significant differences in their immunocompetence and interaction with allogeneic PBMCs, differences may be crucial for the outcome of cell therapy.
Collapse
|
33
|
Harrop JS, Hashimoto R, Norvell D, Raich A, Aarabi B, Grossman RG, Guest JD, Tator CH, Chapman J, Fehlings MG. Evaluation of clinical experience using cell-based therapies in patients with spinal cord injury: a systematic review. J Neurosurg Spine 2012; 17:230-46. [DOI: 10.3171/2012.5.aospine12115] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Object
Using a systematic approach, the authors evaluated the current utilization, safety, and effectiveness of cellular therapies for traumatic spinal cord injuries (SCIs) in humans.
Methods
A systematic search and critical review of the literature published through mid-January 2012 was performed. Articles included in the search were restricted to the English language, studies with at least 10 patients, and those analyzing cellular therapies for traumatic SCI. Citations were evaluated for relevance using a priori criteria, and those that met the inclusion criteria were critically reviewed. Each article was then designated a level of evidence that was developed by the Oxford Centre for Evidence-Based Medicine.
Results
The initial literature search identified 651 relevant articles, which decreased to 350 after excluding case reports and reviews. Evaluation of articles at the title/abstract level, and later at the full-text level, limited the final article set to 12 papers. The following cellular therapies employed in humans with SCI are reviewed: bone marrow mesenchymal and hematopoietic stem cells (8 studies), olfactory ensheathing cells (2 studies), Schwann cells (1 study), and fetal neurogenic tissue (1 study). Overall the quality of the literature was very low, with 3 Grade III levels of evidence and 9 Grade IV studies.
Conclusions
Several different cellular-mediated strategies for adult SCI have been reported to be relatively safe with varying degrees of neurological recovery. However, the literature is of low quality and there is a need for improved preclinical studies and prospective, controlled clinical trials.
Collapse
Affiliation(s)
- James S. Harrop
- 1Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania
| | | | | | | | - Bizhan Aarabi
- 3Department of Neurosurgery, University of Maryland, Baltimore, Maryland
| | | | - James D. Guest
- 5Department of Neurological Surgery, University of Miami, Florida
| | - Charles H. Tator
- 6Department of Neurosurgery, University of Toronto, Ontario, Canada; and
| | - Jens Chapman
- 7Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, Washington
| | | |
Collapse
|
34
|
Sharpe ME, Morton D, Rossi A. Nonclinical safety strategies for stem cell therapies. Toxicol Appl Pharmacol 2012; 262:223-31. [PMID: 22617430 DOI: 10.1016/j.taap.2012.05.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2012] [Revised: 05/02/2012] [Accepted: 05/11/2012] [Indexed: 01/07/2023]
Abstract
Recent breakthroughs in stem cell biology, especially the development of the induced pluripotent stem cell techniques, have generated tremendous enthusiasm and efforts to explore the therapeutic potential of stem cells in regenerative medicine. Stem cell therapies are being considered for the treatment of degenerative diseases, inflammatory conditions, cancer and repair of damaged tissue. The safety of a stem cell therapy depends on many factors including the type of cell therapy, the differentiation status and proliferation capacity of the cells, the route of administration, the intended clinical location, long term survival of the product and/or engraftment, the need for repeated administration, the disease to be treated and the age of the population. Understanding the product profile of the intended therapy is crucial to the development of the nonclinical safety study design.
Collapse
Affiliation(s)
- Michaela E Sharpe
- Investigative Toxicology, Drug Safety Research and Development, Pfizer Ltd, Ramsgate Road, Sandwich, CT13 9NJ, UK.
| | | | | |
Collapse
|
35
|
Schwartz SD, Hubschman JP, Heilwell G, Franco-Cardenas V, Pan CK, Ostrick RM, Mickunas E, Gay R, Klimanskaya I, Lanza R. Embryonic stem cell trials for macular degeneration: a preliminary report. Lancet 2012; 379:713-20. [PMID: 22281388 DOI: 10.1016/s0140-6736(12)60028-2] [Citation(s) in RCA: 963] [Impact Index Per Article: 74.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND It has been 13 years since the discovery of human embryonic stem cells (hESCs). Our report provides the first description of hESC-derived cells transplanted into human patients. METHODS We started two prospective clinical studies to establish the safety and tolerability of subretinal transplantation of hESC-derived retinal pigment epithelium (RPE) in patients with Stargardt's macular dystrophy and dry age-related macular degeneration--the leading cause of blindness in the developed world. Preoperative and postoperative ophthalmic examinations included visual acuity, fluorescein angiography, optical coherence tomography, and visual field testing. These studies are registered with ClinicalTrials.gov, numbers NCT01345006 and NCT01344993. FINDINGS Controlled hESC differentiation resulted in greater than 99% pure RPE. The cells displayed typical RPE behaviour and integrated into the host RPE layer forming mature quiescent monolayers after transplantation in animals. The stage of differentiation substantially affected attachment and survival of the cells in vitro after clinical formulation. Lightly pigmented cells attached and spread in a substantially greater proportion (>90%) than more darkly pigmented cells after culture. After surgery, structural evidence confirmed cells had attached and continued to persist during our study. We did not identify signs of hyperproliferation, abnormal growth, or immune mediated transplant rejection in either patient during the first 4 months. Although there is little agreement between investigators on visual endpoints in patients with low vision, it is encouraging that during the observation period neither patient lost vision. Best corrected visual acuity improved from hand motions to 20/800 (and improved from 0 to 5 letters on the Early Treatment Diabetic Retinopathy Study [ETDRS] visual acuity chart) in the study eye of the patient with Stargardt's macular dystrophy, and vision also seemed to improve in the patient with dry age-related macular degeneration (from 21 ETDRS letters to 28). INTERPRETATION The hESC-derived RPE cells showed no signs of hyperproliferation, tumorigenicity, ectopic tissue formation, or apparent rejection after 4 months. The future therapeutic goal will be to treat patients earlier in the disease processes, potentially increasing the likelihood of photoreceptor and central visual rescue. FUNDING Advanced Cell Technology.
Collapse
Affiliation(s)
- Steven D Schwartz
- Jules Stein Eye Institute Retina Division, Department of Ophthalmology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Affiliation(s)
- R Vawda
- Department of University Health Network, Toronto Western Hospital, Toronto, Canada, ON M5T 2S8
| | - J Wilcox
- Department of University Health Network, Toronto Western Hospital, Toronto, Canada, ON M5T 2S8
| | - MG Fehlings
- Department of University Health Network, Toronto Western Hospital, Toronto, Canada, ON M5T 2S8
| |
Collapse
|
37
|
Abstract
More than 1 million people in the United States live with a spinal cord injury (SCI). Despite medical advances, many patients with SCIs still experience substantial neurological disability, with loss of motor, sensory, and autonomic function. Cell therapy is ideally suited to address the multifactorial nature of the secondary events following SCI. Remarkable advances in our understanding of the pathophysiology of SCI, structural and functional magnetic resonance imaging, image-guided micro-neurosurgical techniques, and transplantable cell biology have enabled the use of cell-based regenerative techniques in the clinic. It is important to note that there are more than a dozen recently completed, ongoing, or recruiting cell therapy clinical trials for SCI that reflect the views of many key stakeholders. The field of regenerative neuroscience has reached a stage in which the clinical trials are scientifically and ethically justified. Although experimental models and analysis methods and techniques continue to evolve, no model will completely replicate the human condition. It is recognized that more work with cervical models of contusive/compressive SCI are required in parallel with clinical trials. It is also important that the clinical translation of advances made through well-established and validated experimental approaches in animal models move forward to meet the compelling needs of individuals with SCI and to advance the field of regenerative neuroscience. However, it is imperative that such efforts at translation be done in the most rigorous and informed fashion to determine safety and possible efficacy, and to provide key information to clinicians and basic scientists, which will allow improvements in regenerative techniques and the validation and refinement of existing preclinical animal models and research approaches. The field of regenerative neuroscience should not be stalled at the animal model stage, but instead the clinical trials need to be focused, safe, and ethical, backed up by a robust, translationally relevant preclinical research strategy.
Collapse
Affiliation(s)
- Michael G. Fehlings
- University Health Network, Toronto Western Hospital, Toronto, ON M5T 2S8 Canada
| | - Reaz Vawda
- University Health Network, Toronto Western Hospital, Toronto, ON M5T 2S8 Canada
| |
Collapse
|
38
|
Watson RA, Yeung TM. What is the potential of oligodendrocyte progenitor cells to successfully treat human spinal cord injury? BMC Neurol 2011; 11:113. [PMID: 21943254 PMCID: PMC3189870 DOI: 10.1186/1471-2377-11-113] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 09/23/2011] [Indexed: 12/28/2022] Open
Abstract
Background Spinal cord injury is a serious and debilitating condition, affecting millions of people worldwide. Long seen as a permanent injury, recent advances in stem cell research have brought closer the possibility of repairing the spinal cord. One such approach involves injecting oligodendrocyte progenitor cells, derived from human embryonic stem cells, into the injured spinal cord in the hope that they will initiate repair. A phase I clinical trial of this therapy was started in mid 2010 and is currently underway. Discussion The theory underlying this approach is that these myelinating progenitors will phenotypically replace myelin lost during injury whilst helping to promote a repair environment in the lesion. However, the importance of demyelination in the pathogenesis of human spinal cord injury is a contentious issue and a body of literature suggests that it is only a minor factor in the overall injury process. Summary This review examines the validity of the theory underpinning the on-going clinical trial as well as analysing published data from animal models and finally discussing issues surrounding safety and purity in order to assess the potential of this approach to successfully treat acute human spinal cord injury.
Collapse
Affiliation(s)
- Robert A Watson
- Green Templeton College, Woodstock Road, Oxford, OX2 6HG, UK.
| | | |
Collapse
|
39
|
Pera MF. Safely modulating the immune system in regenerative medicine. Cell Stem Cell 2011; 8:246-7. [PMID: 21362563 DOI: 10.1016/j.stem.2011.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The destruction of pluripotent stem cell-derived grafts by the host immune system presents a significant barrier to clinical translation of cell therapies. Pearl et al. (2011) report in this issue of Cell Stem Cell that a brief, nontoxic immunosuppressive regimen, achieved by blockade of leucocyte costimulatory pathways, may overcome this problem.
Collapse
Affiliation(s)
- Martin F Pera
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
40
|
Goldring CEP, Duffy PA, Benvenisty N, Andrews PW, Ben-David U, Eakins R, French N, Hanley NA, Kelly L, Kitteringham NR, Kurth J, Ladenheim D, Laverty H, McBlane J, Narayanan G, Patel S, Reinhardt J, Rossi A, Sharpe M, Park BK. Assessing the safety of stem cell therapeutics. Cell Stem Cell 2011; 8:618-28. [PMID: 21624806 DOI: 10.1016/j.stem.2011.05.012] [Citation(s) in RCA: 162] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Unprecedented developments in stem cell research herald a new era of hope and expectation for novel therapies. However, they also present a major challenge for regulators since safety assessment criteria, designed for conventional agents, are largely inappropriate for cell-based therapies. This article aims to set out the safety issues pertaining to novel stem cell-derived treatments, to identify knowledge gaps that require further research, and to suggest a roadmap for developing safety assessment criteria. It is essential that regulators, pharmaceutical providers, and safety scientists work together to frame new safety guidelines, based on "acceptable risk," so that patients are adequately protected but the safety "bar" is not set so high that exciting new treatments are lost.
Collapse
Affiliation(s)
- Chris E P Goldring
- MRC Centre for Drug Safety Science, Division of Molecular & Clinical Pharmacology, The Institute of Translational Medicine, The University of Liverpool, Liverpool L69 3GE, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Kadereit S, Trounson A. In vitro immunogenicity of undifferentiated pluripotent stem cells (PSC) and derived lineages. Semin Immunopathol 2011; 33:551-62. [DOI: 10.1007/s00281-011-0265-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2010] [Accepted: 03/16/2011] [Indexed: 01/19/2023]
|
42
|
Molecular and cellular characterization of expanded and cryopreserved human limbal epithelial stem cells reveal unique immunological properties. Exp Eye Res 2011; 92:47-56. [DOI: 10.1016/j.exer.2010.11.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 10/15/2010] [Accepted: 11/01/2010] [Indexed: 12/24/2022]
|
43
|
Ichim TE, Solano F, Lara F, Paris E, Ugalde F, Rodriguez JP, Minev B, Bogin V, Ramos F, Woods EJ, Murphy MP, Patel AN, Harman RJ, Riordan NH. Feasibility of combination allogeneic stem cell therapy for spinal cord injury: a case report. Int Arch Med 2010; 3:30. [PMID: 21070647 PMCID: PMC2989319 DOI: 10.1186/1755-7682-3-30] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Accepted: 11/11/2010] [Indexed: 12/13/2022] Open
Abstract
Cellular therapy for spinal cord injury (SCI) is overviewed focusing on bone marrow mononuclear cells, olfactory ensheathing cells, and mesenchymal stem cells. A case is made for the possibility of combining cell types, as well as for allogeneic use. We report the case of 29 year old male who suffered a crush fracture of the L1 vertebral body, lacking lower sensorimotor function, being a score A on the ASIA scale. Stem cell therapy comprised of intrathecal administration of allogeneic umbilical cord blood ex-vivo expanded CD34 and umbilical cord matrix MSC was performed 5 months, 8 months, and 14 months after injury. Cell administration was well tolerated with no adverse effects observed. Neuropathic pain subsided from intermittent 10/10 to once a week 3/10 VAS. Recovery of muscle, bowel and sexual function was noted, along with a decrease in ASIA score to "D". This case supports further investigation into allogeneic-based stem cell therapies for SCI.
Collapse
|
44
|
Zhang N, Wimmer J, Qian SJ, Chen WS. Stem Cells: Current Approach and Future Prospects in Spinal Cord Injury Repair. Anat Rec (Hoboken) 2009; 293:519-30. [DOI: 10.1002/ar.21025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Tseng SY, Nishimoto KP, Silk KM, Majumdar AS, Dawes GN, Waldmann H, Fairchild PJ, Lebkowski JS, Reddy A. Generation of immunogenic dendritic cells from human embryonic stem cells without serum and feeder cells. Regen Med 2009; 4:513-26. [PMID: 19580370 DOI: 10.2217/rme.09.25] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM Dendritic cell (DC)-based vaccines have a potential utility for use in the treatment of malignancy. Human embryonic stem cells (hESCs) may provide a more cost-effective and reliable source of DCs for immunotherapy purposes, providing on-demand access for patients. METHOD We developed a protocol to generate DCs from hESCs in vitro in the absence of serum and feeder cells. This protocol uses growth factors bone morphogenetic protein-4, granulocyte macrophage-colony stimulating factor (GM-CSF), stem cell factor and VEGF in serum-free media to generate hESC-derived monocytic cells. These cells are further differentiated to hESC-derived immature DCs with GM-CSF and IL-4, and matured to hESC-derived mature DCs with a maturation cocktail consisting of GM-CSF, TNF-alpha, IL-1beta, IFN-gamma and PGE2. RESULTS This study demonstrates the applicability of our defined differentiation process in generating functional hESC-derived DCs from multiple hESC lines. We show that hESC-derived immature DCs phagocytose, process, and present antigen upon maturation. hESC-derived mature DCs express the maturation marker CD83, produce Th1-directing cytokine IL-12p70, migrate in response to chemokine, and activate both viral and tumor antigen-specific T-cell responses. CONCLUSION We developed a chemically defined system to generate unlimited numbers of DCs from hESCs. Our results demonstrate that hESC-derived DCs generated from this process are immunogenic and have the potential to be used for DC immunotherapy.
Collapse
Affiliation(s)
- Su-Yi Tseng
- Geron Corporation, 230 Constitution Drive, Menlo Park, CA 94025, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lavdas AA, Matsas R. Towards personalized cell-replacement therapies for brain repair. Per Med 2009; 6:293-313. [DOI: 10.2217/pme.09.4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The inability of the CNS to efficiently repair damage caused by trauma and neurodegenerative or demyelinating diseases has underlined the necessity for developing novel therapeutic strategies. Cell transplantation to replace lost neurons and the grafting of myelinating cells to repair demyelinating lesions are promising approaches for treating CNS injuries and demyelination. In this review, we will address the prospects of using stem cells or myelinating glial cells of the PNS, as well as olfactory ensheathing cells, in cell-replacement therapies. The recent generation of induced pluripotent stem cells from adult somatic cells by introduction of three or four genes controlling ‘stemness’ and their subsequent differentiation to desired phenotypes, constitutes a significant advancement towards personalized cell-replacement therapies.
Collapse
Affiliation(s)
- Alexandros A Lavdas
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, 11521 Athens, Greece
| |
Collapse
|
47
|
Ilic D. Industry Update: Latest developments in stem cell research and regenerative medicine. Regen Med 2009. [DOI: 10.2217/rme.09.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Dusko Ilic
- StemLifeLine, Inc., 1300 Industrial Rd. #13, San Carlos, CA 94070, USA
| |
Collapse
|
48
|
Sharp J, Keirstead HS. Stem cell-based cell replacement strategies for the central nervous system. Neurosci Lett 2009; 456:107-11. [PMID: 19429144 DOI: 10.1016/j.neulet.2008.04.106] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 04/15/2008] [Accepted: 04/27/2008] [Indexed: 12/26/2022]
Abstract
During human development, cells of the blastocyst inner cell mass proliferate and give rise to each cell in the human body. It is that potential which focuses intense interest on these stem cells as a substrate for cell-based regenerative medicine. An increased understanding of the interrelation of processes that govern the formation of various cell types will allow for the directed differentiation of stem cells into specified cells or tissues that can ameliorate the effects of disease or damage. Perhaps the most difficult cells and tissues to derive for use in cell replacement strategies are the diverse neurons, glia and complex networks of the central nervous system (CNS). Here we present emerging perspectives on the development of neuronal and glial cells from stem cells for clinical application to CNS diseases and injury.
Collapse
Affiliation(s)
- Jason Sharp
- Reeve-Irvine Research Center, Sue and Bill Gross Stem Cell Research Center, Department of Anatomy & Neurobiology, School of Medicine, 2111 Gillespie Neuroscience Research Facility, University of California at Irvine, Irvine, CA 92697-4292, United States
| | | |
Collapse
|
49
|
Unger C, Skottman H, Blomberg P, Dilber MS, Hovatta O. Good manufacturing practice and clinical-grade human embryonic stem cell lines. Hum Mol Genet 2008; 17:R48-53. [PMID: 18632697 DOI: 10.1093/hmg/ddn079] [Citation(s) in RCA: 160] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Human embryonic stem cell (hESC) lines, after directed differentiation, hold the greatest potential for cell transplantation treatment in many severe diseases. Good manufacturing practice (GMP) quality, defined by both the European Medicines Agency and the Food and Drug Administration, is a requirement for clinical-grade cells, offering optimal defined quality and safety in cell transplantation. Using animal substance-free culture media, feeder cells or feeder-free matrix in derivation, passaging, expansion and cryopreservation procedures, immune reactions against animal proteins in the cells, and infection risk caused by animal microbes can be avoided. It is also possible to apply GMP to animal components if no better options are available. In recent production of GMP-quality hESC lines, feeder cells had been cultured in fetal bovine serum, and the medium supplemented with an animal protein containing a serum replacement component. Using embryos cultured in a GMP laboratory, isolating the inner cell mass mechanically, deriving lines on human feeder cells originally cultured in xeno-free medium in a GMP laboratory, and using xeno-free media for derivation and culture of hESC lines themselves, GMP-quality xeno-free hESC lines could be established today. Human serum is a xeno-free component available today, but many chemically defined media are under development.
Collapse
Affiliation(s)
- Christian Unger
- Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, SE-14186 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
50
|
Abstract
✓ After the successful isolation of human embryonic stem cells in 1998, ethics and policy debates centered on the moral status of the embryo—whether the 2- to 4-day-old blastocyst is a person, and whether we should protect it at all costs. As the research has moved quickly forward, however, new questions have emerged for the study of stem cell ethics, law, and policy. Powerful new lines made without eggs or embryos have recently been reported, the intellectual property and regulatory environment is uncertain, and clinical trials using adult stem cells and cells derived from embryonic stem cells are about to commence. The new landscape of ethics, law, and policy is discussed in the context of these developments, with an emphasis on the evaluation of risks and benefits for first-in-human clinical studies.
Collapse
|