1
|
Farid MF, Abouelela YS, Yasin NAE, Al-Mokaddem AK, Prince A, Ibrahim MA, Rizk H. Laser-activated autologous adipose tissue-derived stromal vascular fraction restores spinal cord architecture and function in multiple sclerosis cat model. Stem Cell Res Ther 2023; 14:6. [PMID: 36627662 PMCID: PMC9832640 DOI: 10.1186/s13287-022-03222-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/14/2022] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Multiple sclerosis (MS) is the most frequent non-traumatic neurological debilitating disease among young adults with no cure. Over recent decades, efforts to treat neurodegenerative diseases have shifted to regenerative cell therapy. Adipose tissue-derived stromal vascular fraction (SVF) comprises a heterogeneous cell population, considered an easily accessible source of MSCs with therapeutic potential in autoimmune diseases. This study aimed to assess the regenerative capacity of low-level laser-activated SVF in an MS cat model. METHODS Fifteen adult Persian cats were used in this study: Group I (control negative group, normal cats), Group II (EB-treated group, induced for MS by ethidium bromide (EB) intrathecal injection), and Group III (SVF co-treated group, induced for MS then treated with SVF on day 14 post-induction). The SVF was obtained after digesting the adipose tissue with collagenase type I and injecting it intrathecal through the foramen magnum. RESULTS The results showed that the pelvic limb's weight-bearing locomotion activity was significantly (P ≤ 0.05) recovered in Group III, and the Basso, Beattie, and Bresnahan (BBB) scores of hindlimb locomotion were significantly higher in Group III (14 ± 0.44) than Group II (4 ± 0.31). The lesion's extent and intensity were reduced in the magnetic resonance imaging (MRI) of Group III. Besides, the same group showed a significant increase in the expression of neurotrophic factors: BDNF, SDF and NGF (0.61 ± 0.01, 0.51 ± 0.01 and 0.67 ± 0.01, respectively) compared with Group II (0.33 ± 0.01, 0.36 ± 0.006 and 0.2 ± 0.01, respectively). Furthermore, SVF co-treated group revealed a significant (P ≤ 0.05) increase in oligodendrocyte transcription factor (Olig2) and myelin basic protein (4 ± 0.35 and 6 ± 0.45, respectively) that was decreased in group II (1.8 ± 0.22 and 2.9 ± 0.20, respectively). Moreover, group III showed a significant (P ≤ 0.05) reduction in Bax and glial fibrillary acidic protein (4 ± 0.53 and 3.8 ± 0.52, respectively) as compared with group II (10.7 ± 0.49 and 8.7 ± 0.78, respectively). The transmission electron microscopy demonstrated regular more compact, and markedly (P ≤ 0.05) thicker myelin sheaths (mm) in Group III (0.3 ± 0.006) as compared with group II (0.1 ± 0.004). Based on our results, the SVF co-treated group revealed remyelination and regeneration capacity with a reduction in apoptosis and axonal degeneration. CONCLUSION SVF is considered an easy, valuable, and promising therapeutic approach for treating spinal cord injuries, particularly MS.
Collapse
Affiliation(s)
- Mariam F. Farid
- grid.7776.10000 0004 0639 9286Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211 Egypt
| | - Yara S. Abouelela
- grid.7776.10000 0004 0639 9286Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211 Egypt
| | - Noha A. E. Yasin
- grid.7776.10000 0004 0639 9286Department of Cytology and Histology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Asmaa K. Al-Mokaddem
- grid.7776.10000 0004 0639 9286Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Abdelbary Prince
- grid.7776.10000 0004 0639 9286Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt ,grid.511523.10000 0004 7532 2290Department of Biomedical Research, Armed Forces College of Medicine, Cairo, 12211 Egypt
| | - Marwa A. Ibrahim
- grid.7776.10000 0004 0639 9286Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Hamdy Rizk
- grid.7776.10000 0004 0639 9286Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza, 12211 Egypt
| |
Collapse
|
2
|
Farid MF, S Abouelela Y, Rizk H. Stem cell treatment trials of spinal cord injuries in animals. Auton Neurosci 2022; 238:102932. [PMID: 35016045 DOI: 10.1016/j.autneu.2021.102932] [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: 09/18/2021] [Revised: 11/01/2021] [Accepted: 12/23/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Spinal cord injury (SCI) is a serious neurological spinal cord damage that resulted in the loss of temporary or permanent function. However, there are even now no effective therapies for it. So, a new medical promising therapeutic hotspot over the previous decades appeared which was (Stem cell (SC) cure of SCI). Otherwise, animal models are considered in preclinical research as a model for humans to trial a potential new treatment. METHODOLOGY Following articles were saved from different databases (PubMed, Google scholar, Egyptian knowledge bank, Elsevier, Medline, Embase, ProQuest, BMC) on the last two decades, and data were obtained then analyzed. RESULTS This review discusses the type and grading of SCI. As well as different types of stem cells therapy for SCI, including mesenchymal stem cells (MSCs), neural stem cells (NSCs), hematopoietic stem cells (HSCs), induced pluripotent stem cells (iPSCs), and embryonic stem cells (ESCs). The review focuses on the transplantation pathways, clinical evaluation, and clinical signs of different types of SC on different animal models which are summarized in tables to give an easy to reach. CONCLUSION Pharmacological and physiotherapy have limited regenerative power in comparison with stem cells medication in the treatment of SCI. Among several sources of cell therapies, mesenchymal stromal/stem cell (MSC) one is being progressively developed as a trusted important energetic way to repair and regenerate. Finally, a wide-ranged animal models have been condensed that helped in human clinical trial therapies.
Collapse
Affiliation(s)
- Mariam F Farid
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| | - Yara S Abouelela
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt.
| | - Hamdy Rizk
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt
| |
Collapse
|
3
|
Richard SA, Sackey M. Elucidating the Pivotal Neuroimmunomodulation of Stem Cells in Spinal Cord Injury Repair. Stem Cells Int 2021; 2021:9230866. [PMID: 34341666 PMCID: PMC8325586 DOI: 10.1155/2021/9230866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/03/2021] [Accepted: 07/17/2021] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a distressing incident with abrupt onset of the motor as well as sensory dysfunction, and most often, the injury occurs as result of high-energy or velocity accidents as well as contact sports and falls in the elderly. The key challenges associated with nerve repair are the lack of self-repair as well as neurotrophic factors and primary and secondary neuronal apoptosis, as well as factors that prevent the regeneration of axons locally. Neurons that survive the initial traumatic damage may be lost due to pathogenic activities like neuroinflammation and apoptosis. Implanted stem cells are capable of differentiating into neural cells that replace injured cells as well as offer local neurotrophic factors that aid neuroprotection, immunomodulation, axonal sprouting, axonal regeneration, and remyelination. At the microenvironment of SCI, stem cells are capable of producing growth factors like brain-derived neurotrophic factor and nerve growth factor which triggers neuronal survival as well as axonal regrowth. Although stem cells have proven to be of therapeutic value in SCI, the major disadvantage of some of the cell types is the risk for tumorigenicity due to the contamination of undifferentiated cells prior to transplantation. Local administration of stem cells via either direct cellular injection into the spinal cord parenchyma or intrathecal administration into the subarachnoid space is currently the best transplantation modality for stem cells during SCI.
Collapse
Affiliation(s)
- Seidu A. Richard
- Department of Medicine, Princefield University, P.O. Box MA128, Ho, Ghana
| | - Marian Sackey
- Department of Pharmacy, Ho Teaching Hospital, P.O. Box MA-374, Ho, Ghana
| |
Collapse
|
4
|
Wang C, Xu H, Liu C, Peng Z, Min R, Zhang Z, Li J, Jin Y, Wang Y, Li Z, Guo J, Zhu L. CaO 2/gelatin oxygen slow-releasing microspheres facilitate tissue engineering efficiency for the osteonecrosis of femoral head by enhancing the angiogenesis and survival of grafted bone marrow mesenchymal stem cells. Biomater Sci 2021; 9:3005-3018. [PMID: 33651043 DOI: 10.1039/d0bm02071k] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The osteonecrosis of femoral head (ONFH), a common refractory disease, is still not fully understood today. Hypoxia caused by ischemia is not only an important pathogenic factor but also a critical challenge for the survival of seed cells in the tissue engineering therapy of ONFH. To explore an efficient strategy to treat ONFH by targeting hypoxia, newly designed CaO2/gelatin microspheres were composited with 3D printed polycaprolactone/nano-hydroxyapatite (PCL/nHA) porous scaffold, sodium alginate/gelatin hydrogel, and bone marrow mesenchymal stem cells (BMSCs) to develop a novel tissue engineering scaffold and then transplanted into the core depression area of the ONFH rabbit model. The current data demonstrated that CaO2/gelatin microspheres can constantly release oxygen for 19 days. In vitro assays with BMSCs illustrated that scaffolds have high biocompatibility and are favorable for cell proliferation in extreme hypoxia (1% O2). The in vivo study demonstrated that the transplanted scaffold with oxygen-generating microspheres significantly enhanced the osteogenic and angiogenic effects compared to the scaffold without microspheres. Further assessments revealed that microspheres in the scaffold can reduce the local cell apoptosis and enhance the survival of grafted cells in the host. Collectively, the present study developed a novel oxygen slow-releasing composite scaffold, which can facilitate tissue engineering efficiency for treating the osteonecrosis of the femoral head by enhancing the angiogenesis and survival of grafted stem cells.
Collapse
Affiliation(s)
- Chengqiang Wang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Haixia Xu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Chun Liu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Ziyue Peng
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Ruoxing Min
- Department of Ultrasound Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhiming Zhang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China. and Department of Orthopedics, Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jianjun Li
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Yanglei Jin
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Yihan Wang
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Zhihao Li
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Jiasong Guo
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China. and Department of Histology and Embryology, Southern Medical University, Guangzhou 510515, China and Key Laboratory of Tissue Construction and Detection of Guangdong Province, Guangzhou 510515, China and Institute of Bone Biology, Academy of Orthopedics, Guangzhou 510665, Guangdong Province, China and Key Laboratory of Mental Health of the Ministry of Education; Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangdong Province Key Laboratory of Psychiatric Disorders, Guangzhou 510515, China and Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou 510530, China
| | - Lixin Zhu
- Department of Spinal Surgery, Orthopedic Medical Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| |
Collapse
|
5
|
Amorim RM, Clark KC, Walker NJ, Kumar P, Herout K, Borjesson DL, Wang A. Placenta-derived multipotent mesenchymal stromal cells: a promising potential cell-based therapy for canine inflammatory brain disease. Stem Cell Res Ther 2020; 11:304. [PMID: 32698861 PMCID: PMC7374910 DOI: 10.1186/s13287-020-01799-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 06/25/2020] [Accepted: 07/01/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Canine inflammatory brain disease (IBD) is a severe inflammatory disorder characterized by infiltration of activated immune cell subsets into the brain and spinal cord. Multipotent mesenchymal stromal cells (MSCs) are a promising therapy for IBD, based on their potent pro-angiogenic, neuroprotective, and immunomodulatory properties. The aims of this study were to compare the immunomodulatory attributes of canine adipose-derived MSCs (ASCs) and placenta-derived MSCs (PMSCs) in vitro. These data will serve as potency information to help inform the optimal MSC cell source to treat naturally occurring canine IBD. METHODS Indoleamine 2,3 dioxygenase (IDO) activity and prostaglandin E2 (PGE2) concentration at baseline and after stimulation with interferon gamma (IFNγ) and/or tumor necrosis factor alpha (TNFα) were measured from canine ASC and PMSC cultures. Leukocyte suppression assays (LSAs) were performed to compare the ability of ASCs and PMSCs to inhibit activated peripheral blood mononuclear cell (PBMC) proliferation. IDO activity and PGE2; interleukin (IL)-2, IL-6, and IL-8; TNFα; and vascular endothelial growth factor (VEGF) concentrations were also measured from co-culture supernatants. Cell cycle analysis was performed to determine how ASCs and PMSCs altered lymphocyte proliferation. RESULTS Activated canine MSCs from both tissue sources secreted high concentrations of IDO and PGE2, after direct stimulation with IFNγ and TNFα, or indirect stimulation by activated PBMCs. Both ASCs and PMSCs inhibited activated PBMC proliferation in LSA assays; however, PMSCs inhibited PBMC proliferation significantly more than ASCs. Blocking PGE2 and IDO in LSA assays determined that PGE2 is important only for ASC inhibition of PBMC proliferation. Activated ASCs increased IL-6 and VEGF secretion and decreased TNFα secretion, while activated PMSCs increased IL-6, IL-8, and VEGF secretion. ASCs inhibited lymphocyte proliferation via cell cycle arrest in the G0/G1 and PMSCs inhibited lymphocyte proliferation via induction of lymphocyte apoptosis. CONCLUSION Our results demonstrate that ASCs and PMSCs have substantial in vitro potential as a cell-based therapy for IBD; however, PMSCs more potently inhibited lymphocyte proliferation by inducing apoptosis of activated lymphocytes. These data suggest that the mechanism by which ASCs and PMSCs downregulate PBMC proliferation differs. Additional studies may elucidate additional mechanisms by which canine MSCs modulate neuroinflammatory responses.
Collapse
Affiliation(s)
- Rogério Martins Amorim
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
- Department of Veterinary Clinics, São Paulo State University “Julio de Mesquita Filho” – UNESP, Botucatu, SP Brazil
| | - Kaitlin C. Clark
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA 95817 USA
- Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals Pediatric Research Center, Northern California, Sacramento, CA USA
| | - Naomi J. Walker
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Priyadarsini Kumar
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA 95817 USA
- Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals Pediatric Research Center, Northern California, Sacramento, CA USA
| | - Kyle Herout
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA 95817 USA
| | - Dori L. Borjesson
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
| | - Aijun Wang
- Veterinary Institute for Regenerative Cures and Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA USA
- Surgical Bioengineering Laboratory, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA 95817 USA
- Institute for Pediatric Regenerative Medicine (IPRM), Shriners Hospitals Pediatric Research Center, Northern California, Sacramento, CA USA
- Department of Biomedical Engineering, University of California, Davis, CA USA
| |
Collapse
|
6
|
Wang JW, Qiu YR, Fu Y, Liu J, He ZJ, Huang ZT. Transplantation with hypoxia-preconditioned mesenchymal stem cells suppresses brain injury caused by cardiac arrest-induced global cerebral ischemia in rats. J Neurosci Res 2017; 95:2059-2070. [PMID: 28186348 DOI: 10.1002/jnr.24025] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 12/22/2016] [Accepted: 12/28/2016] [Indexed: 12/16/2022]
Abstract
Cardiac arrest-induced global cerebral ischemia is a main cause of neurological dysfunction in emergency medicine. Transplantation with bone marrow mesenchymal stem cells (MSCs) has been used in stroke models to repair the ischemic brain injury, but it is little studied in models with global cerebral ischemia. In the present study, a hypoxia precondition was used to improve the efficacy of MSC transplantation, given the low survival and migration rates and limited differentiation capacities of MSCs. We found that hypoxia can increase the expansion and migration of MSCs by activating the PI3K/AKT and hypoxia-inducible factor-1α/CXC chemokine receptor-4 pathways. By using a cardiac arrest-induced global cerebral ischemic model in rats, we found that transplantation of hypoxia-preconditioned MSCs promoted the migration and integration of MSCs and decreased neuronal death and inflammation in the ischemic cortex. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Ji-Wen Wang
- Department of Intensive Care Unit, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guang Zhou, China.,Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-Sen University, Guang Zhou, China
| | - Yu-Ru Qiu
- Department of Intensive Care Unit, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guang Zhou, China
| | - Yue Fu
- Department of Emergency Medicine, First People's Hospital of Fo Shan, Fo Shan, China
| | - Jun Liu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guang Zhou, China
| | - Zhi-Jie He
- Department of Intensive Care Unit, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guang Zhou, China
| | - Zi-Tong Huang
- Institute of Cardiopulmonary Cerebral Resuscitation, Sun Yat-Sen University, Guang Zhou, China.,Department of Emergency Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guang Zhou, China
| |
Collapse
|
7
|
Shichinohe H, Houkin K. Cell Therapy for Stroke: Review of Previous Clinical Trials and Introduction of Our New Trials. Neurol Med Chir (Tokyo) 2016; 56:592-596. [PMID: 27302193 PMCID: PMC5066079 DOI: 10.2176/nmc.st.2016-0087] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stroke is still a leading cause of death and disability, and despite intensive research, few treatment options exist. A recent breakthrough in cell therapy is expected to reverse the neurological sequelae of stroke. Although some pioneer studies on the use of cell therapy for the treatment of stroke have been reported, certain problems still remain unsolved. We investigated the use of autologous bone marrow stromal cell (BMSC) transplantation for the treatment of stroke, to develop it as the next-generation cell therapy. In this study, we introduce the preparation of a new clinical trial, the Research on Advanced Intervention using Novel Bone marrow stem cell (RAINBOW) study. The trial will start in 2016, and we hope that it will not only be helpful for treating patients but also for clarifying the therapeutic mechanisms. Moreover, we review stem cell therapeutics as an emerging paradigm in stroke (STEPS) and the guidelines for the development of cell therapy for stroke in the United States as well as introduce the development of new guidelines in Japan. These guidelines are expected to encourage the development of cell therapy for stroke management.
Collapse
Affiliation(s)
- Hideo Shichinohe
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine
| | | |
Collapse
|
8
|
Li R. Stem cell transplantation for treating Parkinson's disease: Literature analysis based on the Web of Science. Neural Regen Res 2015; 7:1272-9. [PMID: 25709626 PMCID: PMC4336963 DOI: 10.3969/j.issn.1673-5374.2012.16.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 04/23/2012] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE: To identify global research trends of stem cell transplantation for treating Parkinson's disease using a bibliometric analysis of the Web of Science. DATA RETRIEVAL: We performed a bibliometric analysis of data retrievals for stem cell transplantation for treating Parkinson's disease from 2002 to 2011 using the Web of Science. SELECTION CRITERIA: Inclusion criteria: (a) peer-reviewed articles on stem cell transplantation for treating Parkinson's disease which were published and indexed in the Web of Science; (b) type of articles: original research articles, reviews, meeting abstracts, proceedings papers, book chapters, editorial material and news items; (c) year of publication: 2002–2011. Exclusion criteria: (a) articles that required manual searching or telephone access; (b) we excluded documents that were not published in the public domain; (c) we excluded a number of corrected papers from the total number of articles. MAIN OUTCOME MEASURES: (1) Type of literature; (2) annual publication output; (3) distribution according to journals; (4) distribution according to subject areas; (5) distribution according to country; (6) distribution according to institution; (7) comparison of countries that published the most papers on stem cell transplantation from different cell sources for treating Parkinson's disease; (8) comparison of institutions that published the most papers on stem cell transplantation from different cell sources for treating Parkinson's disease in the Web of Science from 2002 to 2011; (9) comparison of studies on stem cell transplantation from different cell sources for treating Parkinson's disease RESULTS: In total, 1 062 studies on stem cell transplantation for treating Parkinson's disease appeared in the Web of Science from 2002 to 2011, almost one third of which were from American authors and institutes. The number of studies on stem cell transplantation for treating Parkinson's disease had gradually increased over the past 10 years. Papers on stem cell transplantation for treating Parkinson's disease appeared in journals such as Stem Cells and Experimental Neurology. Although the United States published more articles addressing neural stem cell and embryonic stem cell transplantation for treating Parkinson's disease, China ranked first for articles published on bone marrow mesenchymal stem cell transplantation for treating Parkinson's disease. CONCLUSION: From our analysis of the literature and research trends, we found that stem cell transplantation for treating Parkinson's disease may offer further benefits in regenerative medicine.
Collapse
Affiliation(s)
- Runhui Li
- Department of Neurology, Fengtian Hospital of Shenyang Medical College, Shenyang 110024, Liaoning Province, China
| |
Collapse
|
9
|
Wakao S, Akashi H, Kushida Y, Dezawa M. Muse cells, newly found non-tumorigenic pluripotent stem cells, reside in human mesenchymal tissues. Pathol Int 2014; 64:1-9. [PMID: 24471964 DOI: 10.1111/pin.12129] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 12/13/2013] [Indexed: 01/09/2023]
Abstract
Mesenchymal stem cells (MSCs) have been presumed to include a subpopulation of pluripotent-like cells as they differentiate not only into the same mesodermal-lineage cells but also into ectodermal- and endodermal-lineage cells and exert tissue regenerative effects in a wide variety of tissues. A novel type of pluripotent stem cell, Multilineage-differentiating stress enduring (Muse) cells, was recently discovered in mesenchymal tissues such as the bone marrow, adipose tissue, dermis and connective tissue of organs, as well as in cultured fibroblasts and bone marrow-MSCs. Muse cells are able to differentiate into all three germ layers from a single cell and to self-renew, and yet exhibit non-tumorigenic and low telomerase activities. They can migrate to and target damaged sites in vivo, spontaneously differentiate into cells compatible with the targeted tissue, and contribute to tissue repair. Thus, Muse cells may account for the wide variety of differentiation abilities and tissue repair effects that have been observed in MSCs. Muse cells are unique in that they are pluripotent stem cells that belong in the living body, and are thus assumed to play an important role in 'regenerative homeostasis' in vivo.
Collapse
Affiliation(s)
- Shohei Wakao
- Department of Stem Cell Biology and Histology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | |
Collapse
|
10
|
Tse KH, Novikov LN, Wiberg M, Kingham PJ. Intrinsic mechanisms underlying the neurotrophic activity of adipose derived stem cells. Exp Cell Res 2014; 331:142-151. [PMID: 25193075 DOI: 10.1016/j.yexcr.2014.08.034] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 08/24/2014] [Indexed: 01/14/2023]
Abstract
Adipose derived stem cells (ADSC) can be differentiated into Schwann cell-like cells which enhance nerve function and regeneration. However, the signalling mechanisms underlying the neurotrophic potential of ADSC remain largely unknown. In this study, we hypothesised that ADSC, upon stimulation with a combination of growth factors, could rapidly produce brain derived neurotrophic factor (BDNF) with a similar molecular mechanism to that functioning in the nervous system. Within 48 h of stimulation, ADSC demonstrated potent neurotrophic effects on dorsal root ganglion neurons, at a magnitude equivalent to that of the longer term differentiated Schwann cell-like cells. Stimulated ADSC showed rapid up-regulation of the neuronal activity dependent promoter BDNF exon IV along with an augmented expression of full length protein encoding BDNF exon IX. BDNF protein was secreted at a concentration similar to that produced by differentiated Schwann cell-like cells. Stimulation also activated the BDNF expression gating transcription factor, cAMP responsive element binding (CREB) protein. However, blocking phosphorylation of CREB with the protein kinase A small molecule inhibitor H89 did not suppress secretion of BDNF protein. These results suggest rapid BDNF production in ADSC is mediated through multiple compensatory pathways independent of, or in addition to, the CREB neuronal activation cascade.
Collapse
Affiliation(s)
- Kai-Hei Tse
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, SE-901 87 Umeå, Sweden
| | - Lev N Novikov
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, SE-901 87 Umeå, Sweden
| | - Mikael Wiberg
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, SE-901 87 Umeå, Sweden; Department of Surgical and Perioperative Sciences, Section of Hand & Plastic Surgery, Umeå University, Sweden
| | - Paul J Kingham
- Department of Integrative Medical Biology, Section of Anatomy, Umeå University, SE-901 87 Umeå, Sweden.
| |
Collapse
|
11
|
Genetically modified mesenchymal stem cells (MSCs) promote axonal regeneration and prevent hypersensitivity after spinal cord injury. Exp Neurol 2013; 248:369-80. [PMID: 23856436 DOI: 10.1016/j.expneurol.2013.06.028] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 06/10/2013] [Accepted: 06/28/2013] [Indexed: 12/14/2022]
Abstract
Neurotrophins and the transplantation of bone marrow-derived stromal cells (MSCs) are both candidate therapies targeting spinal cord injury (SCI). While some studies have suggested the ability of MSCs to transdifferentiate into neural cells, other SCI studies have proposed anti-inflammatory and other mechanisms underlying established beneficial effects. We grafted rat MSCs genetically modified to express MNTS1, a multineurotrophin that binds TrkA, TrkB and TrkC, and p75(NTR) receptors or MSC-MNTS1/p75(-) that binds mainly to the Trk receptors. Seven days after contusive SCI, PBS-only, GFP-MSC, MSC-MNTS1/GFP or MSC-MNTS1/p75(-)/GFP were delivered into the injury epicenter. All transplanted groups showed reduced inflammation and cystic cavity size compared to control SCI rats. Interestingly, transplantation of the MSC-MNTS1 and MSC-MNTS1/p75(-), but not the naïve MSCs, enhanced axonal growth and significantly prevented cutaneous hypersensitivity after SCI. Moreover, transplantation of MSC-MNTS1/p75(-) promoted angiogenesis and modified glial scar formation. These findings suggest that MSCs transduced with a multineurotrophin are effective in promoting cell growth and improving sensory function after SCI. These novel data also provide insight into the neurotrophin-receptor dependent mechanisms through which cellular transplantation leads to functional improvement after experimental SCI.
Collapse
|
12
|
Lin W, Li M, Li Y, Sun X, Li X, Yang F, Huang Y, Wang X. Bone marrow stromal cells promote neurite outgrowth of spinal motor neurons by means of neurotrophic factors in vitro. Neurol Sci 2013; 35:449-57. [PMID: 23832111 DOI: 10.1007/s10072-013-1490-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2012] [Accepted: 06/24/2013] [Indexed: 12/13/2022]
Abstract
Transplantation of bone marrow stromal cells (BMSCs) into spinal cord injury models has shown significant neural function recovery; however, the underlying mechanisms have not been fully understood. In the present study we examined the effect of BMSCs on neurite outgrowth of spinal motor neuron using an in vitro co-culture system. The ventral horn of the spinal grey matter was harvested from neonatal Sprague-Dawley rats, cultured with BMSCs, and immunostained for neurofilament-200 (NF-200). Neurite outgrowth of spinal motor neurons was measured using Image J software. ELISA was used to quantify neurotrophic factors such as brain-derived neurotrophic factor (BDNF), glial cell line-derived neurotrophic factor (GDNF) and nerve growth factor (NGF) in culture media, and antibodies or exogenous neurotrophic factors were used to block or mimic the effect of BMSCs on neurite outgrowth, respectively. The results showed that neurite outgrowth significantly increased in spinal motor neurons after co-cultured with BMSCs, while the secretion level of BDNF, GDNF and NGF was dramatically elevated in co-culture. However, the neurite outgrowth-promoting effect of BMSCs was found to significantly reduced using antibodies to BDNF, GDNF and NGF. In addition, a fraction of BMSCs was found to exhibit NF-200 immunoreactivity. These results indicated that BMSCs could promote neurite outgrowth of motor neurons by means of neurotrophic factors. The findings of the present study provided new cues for the treatment of spinal cord injury.
Collapse
Affiliation(s)
- Weiwei Lin
- Department of Histology and Embryology, Medical College, Nantong University, 19 Qixiu Road, Nantong, 226001, JS, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Naghdi M, Tiraihi T, Mesbah-Namin SA, Arabkharadmand J, Kazemi H, Taheri T. Improvement of Contused Spinal Cord in Rats by Cholinergic-like Neuron Therapy. IRANIAN RED CRESCENT MEDICAL JOURNAL 2013; 15:127-35. [PMID: 23682324 PMCID: PMC3652499 DOI: 10.5812/ircmj.7653] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 01/08/2013] [Indexed: 12/28/2022]
Abstract
Background Disability in spinal cord injury is an important medical problem, and cell transplantation is considered as an option for the treatment. Objectives The purpose of this study is to use bone marrow stromal cells (BMSCs) derived cholinergic neuron-like cells (CNL) in order to ameliorate the contusion model of spinal cord injury in rats. Materials and Methods The CNLs were produced by pre inducing BMSCs with β-mercaptoethanol (BME) followed by inducing with nerve growth factor (NGF). The cells were immunoreactive to neurofilament 200, NeuN, synaptophysin, synapsin, microtubule associated protein-2 and choline acetyl transferase (ChAT). The CNL were transplanted in contused rats (CR), which were sacrificed after 12 weeks. Results The results showed that BBB test showed an improvement in the CR, while the quantitative analysis showed that the improvement rate was higher in the rats treated with CNL than those treated with BMSCs only or the untreated animals, similar results were noticed in the improvement index. Immunohistochemical analysis of the tissue section prepared from the CR showed that the transplanted cells were engrafted and integrated in the traumatized spinal cord. The morphometric analysis showed that the volume density of the cavity in the CNL treated rats was significantly lower than that of the untreated ones, while the spinal tissue regeneration index was significantly higher. Conclusions The conclusion of the study is that CNL can improve the injured spinal cord.
Collapse
Affiliation(s)
- Majid Naghdi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
| | - Taki Tiraihi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University; Shefa Neuroscience Research Center, Khatam Al-anbia Hospital, Tehran, IR Iran
- Corresponding author: Taki Tiraihi, Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, P.O: 14155-4838, Tehran, IR Iran. Tel: +98-2183553920, Fax: +98-2183553920, E-mail:
| | - Seyed Alireza Mesbah-Namin
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, IR Iran
| | | | - Hadi Kazemi
- Shefa Neuroscience Research Center, Khatam Al-anbia Hospital, Tehran, IR Iran
| | - Taher Taheri
- Shefa Neuroscience Research Center, Khatam Al-anbia Hospital, Tehran, IR Iran
| |
Collapse
|
14
|
Ozdemir M, Attar A, Kuzu I, Ayten M, Ozgencil E, Bozkurt M, Dalva K, Uckan D, Kılıc E, Sancak T, Kanpolat Y, Beksac M. Stem cell therapy in spinal cord injury: in vivo and postmortem tracking of bone marrow mononuclear or mesenchymal stem cells. Stem Cell Rev Rep 2012; 8:953-62. [PMID: 22552878 DOI: 10.1007/s12015-012-9376-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE The aim of this study was to address the question of whether bone marrow-originated mononuclear cells (MNC) or mesenchymal stem cells (MSC) induce neural regeneration when implanted intraspinally. MATERIALS AND METHODS The study design included 4 groups of mice: Group 1, non-traumatized control group; Groups 2, 3 and 4 spinal cord traumatized mice with 1 g force Tator clips, which received intralesionally either no cellular implants (Group 2), luciferase (Luc) (+) MNC (Group 3) or MSC (Group 4) obtained from CMV-Luc or beta-actin Luc donor transgenic mice. Following the surgery until decapitation, periodical radioluminescence imaging (RLI) and Basso Mouse Scale (BMS) evaluations was performed to monitor neural activity. Postmortem immunohistochemical techniques were used to analyze the fate of donor type implanted cells. RESULTS All mice of Groups 3 and 4 showed various degrees of improvement in the BMS scores, whereas there was no change in Groups 1 and 2. The functional improvement was significantly better in Group 4 compared to Group 3 (18 vs 8, p=0.002). The immunohistochemical staining demonstrated GFP(+)Luc(+) neuronal/glial cells that were also positive with one or more of these markers: nestin, myelin associated glycoprotein, microtubule associated protein or myelin oligodendrocyte specific protein, which is considered as indicator of donor type neuronal regeneration. Frequency of donor type neuronal cells; Luc + signals and median BMS scores were observed 48-64% and 68-72%; 44-80%; 8 and 18 within Groups III and IV respectively. DISCUSSION MSCs were more effective than MNC in obtaining neuronal recovery. Substantial but incomplete functional improvement was associated with donor type in vivo imaging signals more frequently than the number of neuronal cells expressing donor markers in spinal cord sections in vitro. Our results are in favor of functional recovery arising from both donor MSC and MNCs, contributing to direct neuronal regeneration and additional indirect mechanisms.
Collapse
Affiliation(s)
- Mevci Ozdemir
- School of Medicine, Department of Neurosurgery, Pamukkale University, 20070, Kinikli, Denizli, Turkey.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Shichinohe H, Kuroda S, Kudo K, Ito M, Kawabori M, Miyamoto M, Nakanishi M, Terae S, Houkin K. Visualization of the Superparamagnetic Iron Oxide (SPIO)-Labeled Bone Marrow Stromal Cells Using a 3.0-T MRI—a Pilot Study for Clinical Testing of Neurotransplantation. Transl Stroke Res 2011; 3:99-106. [DOI: 10.1007/s12975-011-0138-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 11/28/2011] [Accepted: 11/29/2011] [Indexed: 01/19/2023]
|
16
|
Shichinohe H, Kuroda S, Sugiyama T, Ito M, Kawabori M, Nishio M, Takeda Y, Koike T, Houkin K. Biological Features of Human Bone Marrow Stromal Cells (hBMSC) Cultured with Animal Protein-Free Medium—Safety and Efficacy of Clinical Use for Neurotransplantation. Transl Stroke Res 2011; 2:307-15. [DOI: 10.1007/s12975-011-0088-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2011] [Revised: 05/24/2011] [Accepted: 06/03/2011] [Indexed: 01/07/2023]
|
17
|
Kuroda S, Shichinohe H, Houkin K, Iwasaki Y. Autologous bone marrow stromal cell transplantation for central nervous system disorders - recent progress and perspective for clinical application. J Stem Cells Regen Med 2011. [PMID: 24693168 PMCID: PMC3908285 DOI: 10.46582/jsrm.0701002] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
There is increasing evidence that the transplanted BMSC significantly promote functional recovery after CNS damage in the animal models of various kinds of CNS disorders, including cerebral infarct, traumatic brain injury and spinal cord injury. However, there are several shortages of information when considering clinical application of BMSC transplantation for patients with CNS disorders. In this review, therefore, we discuss what we should clarify to establish cell transplantation therapy as the scientifically proven entity in clinical situation and describe our recent works for this purpose. The BMSC have the ability to alter their gene expression profile and phenotype in response to the surrounding circumstances and to protect the neurons by producing some neurotrophic factors. They also promote neurite extension and rebuild the neural circuits in the injured CNS. The BMSC can be expanded in vitro using the animal serum-free medium. Pharmacological modulation may accelerate the in vitro proliferation of the BMSC. Using in vivo optical imaging technique, the transplanted BMSC can non-invasively be tracked in the living animals for at least 8 weeks after transplantation. It is urgent issues to develop clinical imaging technique to track the transplanted cells in the CNS and evaluate the therapeutic significance of BMSC transplantation in order to establish it as a definite therapeutic strategy in clinical situation in the future.
Collapse
Affiliation(s)
- S Kuroda
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine , Sapporo, Japan
| | - H Shichinohe
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine , Sapporo, Japan
| | - K Houkin
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine , Sapporo, Japan
| | - Y Iwasaki
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine , Sapporo, Japan
| |
Collapse
|
18
|
Wu W, Zhao H, Xie B, Liu H, Chen Y, Jiao G, Wang H. Implanted spike wave electric stimulation promotes survival of the bone marrow mesenchymal stem cells and functional recovery in the spinal cord injured rats. Neurosci Lett 2011; 491:73-8. [PMID: 21232582 DOI: 10.1016/j.neulet.2011.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 12/27/2010] [Accepted: 01/04/2011] [Indexed: 12/20/2022]
Abstract
Transplantation of bone marrow-derived mesenchymal stromal cells (BMSCs) into the injured spinal cord may provide therapeutic benefit, but its application is limited by their poor survival and low differentiation rate into neurons. Electrical stimulation (ES) has been reported to promote survival and differentiation of the BMSCs. Therefore we investigated whether implanted spike wave ES could improve survival of BMSCs after transplantation and result in functional improvement in animals with spinal cord injury. Our results showed that the number and ratio of survived BMSCs near the lesion site were significantly increased in the BMSCs+ES-treated group as compared to BMSCs transplantation or ES treatment alone group. Furthermore, results from BBB scales, SSEP and DTI demonstrated a significant improved functional recovery in the BMSCs+ES group. This indicated that implanted spike wave ES could promote the bioactivity of BMSCs and their survival. This represents a new therapeutic potential of the combination of BMSCs transplantation with implanted spike wave ES to treat spinal cord injury.
Collapse
Affiliation(s)
- Wenliang Wu
- Department of Trauma Surgery, Shandong University Qilu Hospital, Wenhuaxi Road 107, Jinan 250012, China
| | | | | | | | | | | | | |
Collapse
|
19
|
Giannoni P, Scaglione S, Daga A, Ilengo C, Cilli M, Quarto R. Short-time survival and engraftment of bone marrow stromal cells in an ectopic model of bone regeneration. Tissue Eng Part A 2010; 16:489-99. [PMID: 19712045 DOI: 10.1089/ten.tea.2009.0041] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
In tissue-engineered applications bone marrow stromal cells (BMSCs) are combined with scaffolds to target bone regeneration; animal models have been devised and the cells' long-term engraftment has been widely studied. However, in regenerated bone, the cell number is severely reduced with respect to the initially seeded BMSCs. This reflects the natural low cellularity of bone but underlines the selectivity of the differentiation processes. In this respect, we evaluated the short-term survival of BMSCs, transduced with the luciferase gene, after implantation of cell-seeded scaffolds in a nude mouse model. Cell proliferation/survival was assessed by bioluminescence imaging: light production was decreased by 30% on the first day, reaching a 50% loss within 48 h. Less than 5% of the initial signal remained after 2 months in vivo. Apoptotic BMSCs were detected within the first 2 days of implantation. Interestingly, the initial frequency of clonogenic progenitors matched the percentage of in vivo surviving cells. Cytokines and inflammation may contribute to the apoptotic onset at the implant milieu. However, preculturing cells with tumor necrosis factor alpha enhanced survival, allowing detection of 8.1% of the seeded BMSCs 2 months after implantation. Thus culturing conditions may reduce the apoptotic overload of seeded osteoprogenitors, strengthening the constructs' osteogenic potential.
Collapse
Affiliation(s)
- Paolo Giannoni
- Stem Cell Laboratory, Advanced Biotechnology Center, Genova, Italy.
| | | | | | | | | | | |
Collapse
|
20
|
Tetzlaff W, Okon EB, Karimi-Abdolrezaee S, Hill CE, Sparling JS, Plemel JR, Plunet WT, Tsai EC, Baptiste D, Smithson LJ, Kawaja MD, Fehlings MG, Kwon BK. A systematic review of cellular transplantation therapies for spinal cord injury. J Neurotrauma 2010; 28:1611-82. [PMID: 20146557 DOI: 10.1089/neu.2009.1177] [Citation(s) in RCA: 419] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cell transplantation therapies have become a major focus in pre-clinical research as a promising strategy for the treatment of spinal cord injury (SCI). In this article, we systematically review the available pre-clinical literature on the most commonly used cell types in order to assess the body of evidence that may support their translation to human SCI patients. These cell types include Schwann cells, olfactory ensheathing glial cells, embryonic and adult neural stem/progenitor cells, fate-restricted neural/glial precursor cells, and bone-marrow stromal cells. Studies were included for review only if they described the transplantation of the cell substrate into an in-vivo model of traumatic SCI, induced either bluntly or sharply. Using these inclusion criteria, 162 studies were identified and reviewed in detail, emphasizing their behavioral effects (although not limiting the scope of the discussion to behavioral effects alone). Significant differences between cells of the same "type" exist based on the species and age of donor, as well as culture conditions and mode of delivery. Many of these studies used cell transplantations in combination with other strategies. The systematic review makes it very apparent that cells derived from rodent sources have been the most extensively studied, while only 19 studies reported the transplantation of human cells, nine of which utilized bone-marrow stromal cells. Similarly, the vast majority of studies have been conducted in rodent models of injury, and few studies have investigated cell transplantation in larger mammals or primates. With respect to the timing of intervention, nearly all of the studies reviewed were conducted with transplantations occurring subacutely and acutely, while chronic treatments were rare and often failed to yield functional benefits.
Collapse
Affiliation(s)
- Wolfram Tetzlaff
- University of British Columbia, ICORD, Vancouver, British Columbia, Canada.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Shichinohe H, Kuroda S, Maruichi K, Osanai T, Sugiyama T, Chiba Y, Yamaguchi A, Iwasaki Y. Bone marrow stromal cells and bone marrow-derived mononuclear cells: Which are suitable as cell source of transplantation for mice infarct brain? Neuropathology 2010; 30:113-22. [DOI: 10.1111/j.1440-1789.2009.01050.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
22
|
Yasuda H, Kuroda S, Shichinohe H, Kamei S, Kawamura R, Iwasaki Y. Effect of biodegradable fibrin scaffold on survival, migration, and differentiation of transplanted bone marrow stromal cells after cortical injury in rats. J Neurosurg 2010; 112:336-44. [DOI: 10.3171/2009.2.jns08495] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Object
In this study the authors' aim was to assess whether fibrin matrix could act as an injectable, valuable scaffold in bone marrow stromal cell (BMSC) transplantation for injured CNS tissue.
Methods
Both clotting time and 3D structure of fibrin matrix were analyzed with various concentrations of fibrinogen and CaCl2. The BMSCs were harvested from green fluorescent protein–transgenic mice and cultured. A cortical lesion was produced in rats by application of a very cold rod to the right cerebral hemisphere. The BMSCs, fibrin matrix, or BMSC–fibrin matrix complex was transplanted into the lesion though a small bur hole 7 days after the insult. Using immunohistochemical analysis, the authors evaluated the survival, migration, and differentiation of the transplanted cells 4 weeks after transplantation.
Results
Based on in vitro observations, the concentrations of fibrinogen and CaCl2 were fixed at 2.5 mg/ml and 2 μM in animal experiments, respectively. Fibrin matrix almost completely disappeared 4 weeks after transplantation. However, immunohistochemical analysis revealed that fibrin matrix exclusively enhanced the retention of the transplanted cells within the lesion, migration toward the lesion boundary zone, and differentiation into the neurons and perivascular cells.
Conclusions
Injectable fibrin matrix enhanced the survival, migration, and differentiation of the BMSCs transplanted into the cortical lesion in rats. The authors believe that it is one of the promising candidates for a potential, minimally invasive scaffold for CNS disorders. The present findings strongly suggest that such a strategy of tissue engineering could be a therapeutic option for CNS regeneration in patients with CNS injuries.
Collapse
Affiliation(s)
- Hiroshi Yasuda
- 1Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo; and
| | - Satoshi Kuroda
- 1Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo; and
| | - Hideo Shichinohe
- 1Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo; and
| | - Shintaro Kamei
- 2The Chemo-Sero-Therapeutic Research Institute, Kumamoto, Japan
| | | | - Yoshinobu Iwasaki
- 1Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo; and
| |
Collapse
|
23
|
Shichinohe H, Kuroda S, Tsuji S, Yamaguchi S, Yano S, Lee JB, Kobayashi H, Kikuchi S, Hida K, Iwasaki Y. Bone marrow stromal cells promote neurite extension in organotypic spinal cord slice: significance for cell transplantation therapy. Neurorehabil Neural Repair 2010; 22:447-57. [PMID: 18780880 DOI: 10.1177/1545968308315596] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Recent reports have indicated that bone marrow stromal cells (BMSCs) have the potential to improve neurological function when transplanted into models of central nervous system (CNS) disorders, including traumatic spinal cord injury. In this study, the authors aimed to clarify the underlying mechanism through which BMSCs supported CNS regeneration in the spinal cord. METHODS The authors topically applied mouse BMSCs expressing green fluorescence protein (0.4-4 x 10(4) cells) on the organotypic spinal cord slice culture prepared from 6-day-old rat pups (n = 17). They were co-cultured for 3 weeks after the slice culture started, and the behavior of the applied BMSCs was serially observed using a fluorescence bioimaging technique. The authors completed a histological analysis at the end of the co-cultures and evaluated the profiles of the cultured BMSCs using microarray and immunocytochemistry techniques. RESULTS The fluorescence bioimaging showed that the BMSCs survived and made a cluster on the slice during the experiments. They also induced a morphological change in the slice within 48 hours of co-culture. Immunohistochemistry analysis showed that the BMSCs promoted a marked neurite extension toward their cluster and some of the BMSCs expressed Tuj-1, an early neuronal marker. Analysis by microarray and immunocytochemistry revealed that BMSCs highly expressed the matrix metalloproteinases (MMPs), stromal cell-derived factor-1, and its specific receptor CXCR4. CONCLUSIONS These findings suggest that the donor BMSCs can support CNS regeneration due to their acquisition of a suitable environment for differentiation and promotion of neurite extension via MMPs and chemokines.
Collapse
Affiliation(s)
- Hideo Shichinohe
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhang HT, Luo J, Sui LS, Ma X, Yan ZJ, Lin JH, Wang YS, Chen YZ, Jiang XD, Xu RX. Effects of differentiated versus undifferentiated adipose tissue-derived stromal cell grafts on functional recovery after spinal cord contusion. Cell Mol Neurobiol 2009; 29:1283-92. [PMID: 19533335 PMCID: PMC11505793 DOI: 10.1007/s10571-009-9424-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2008] [Accepted: 06/03/2009] [Indexed: 02/06/2023]
Abstract
Controversies exist concerning the need for mesenchymal stromal cells (MSCs) to be transdifferentiated prior to their transplantation. In the present study, we compared the results of grafting into the rat contused spinal cord undifferentiated, adipose tissue-derived stromal cells (uADSCs) versus ADSCs induced by two different protocols to form differentiated nervous tissue. Using Basso, Beattie, and Bresnahan scores and grid tests, we found that three cell-treated groups, including uADSCs-treated, dADSCs induced by Protocol 1 (dADSC-P1)-treated, and dADSCs induced by Protocol 2 (dADSC-P2)-treated groups, significantly improved locomotor functional recovery in SCI rats, compared with the saline-treated group. Furthermore, functional recovery was better in the uADSC-treated and dADSC-P2-treated groups than in the dADSC-P1-treated group at week 12 postinjury (P < 0.05 for dADSC-P1 group vs. uADSCs or dADSC-P2 groups). Although both protocols could induce high percentages of cells expressing neural markers in vitro, few BrdU-labeled cells survived at the injury sites in the three cell-treated groups, and only a small percentage of BrdU-positive cells expressed neural markers. On the other hand, the number of NF200-positive axons in the uADSC-treated and dADSC-P2-treated groups was significantly larger than those in the dADSC-P1-treated and saline-treated control groups. Our results indicate that ADSCs are able to differentiate into neural-like cells in vitro and in vivo. However, neural differentiated ADSCs did not result in better functional recovery than undifferentiated ones, following SCI. In vitro neural transdifferentiation of ADSCs might therefore not be a necessary pretransplantation step. Furthermore, cellular replacement or integration might not contribute to the functional recovery of the injured spinal cord.
Collapse
Affiliation(s)
- Hong-Tian Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Jie Luo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Li-Sen Sui
- Department of Neurosurgery, Guangdong Hospital of Traditional Chinese Medicine, 510120 Guangzhou, China
| | - Xu Ma
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Zhong-Jie Yan
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Jian-Hao Lin
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Yu-Sheng Wang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Yi-Zhao Chen
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Xiao-Dan Jiang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
| | - Ru-Xiang Xu
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, 510282 Guangzhou, China
- Institute of Neurosurgery, Key Laboratory on Brain Function Repair and Regeneration of Guangdong, Southern Medical University, 510282 Guangzhou, China
- Department of Neurosurgery, The Military General Hospital of Beijing PLA, 100700 Beijing, China
| |
Collapse
|
25
|
Chiba Y, Kuroda S, Shichinohe H, Hokari M, Osanai T, Maruichi K, Yano S, Hida K, Iwasaki Y. Synergistic effects of bone marrow stromal cells and a Rho kinase (ROCK) inhibitor, Fasudil on axon regeneration in rat spinal cord injury. Neuropathology 2009; 30:241-50. [DOI: 10.1111/j.1440-1789.2009.01077.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
26
|
Combination of bone marrow stromal cell transplantation with mobilization by granulocyte-colony stimulating factor promotes functional recovery after spinal cord transection. Acta Neurochir (Wien) 2009; 151:1483-92. [PMID: 19499175 DOI: 10.1007/s00701-009-0402-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2009] [Accepted: 05/01/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE Spinal cord injury (SCI) results in severe neurological deficit. However, the functional recovery following SCI is very poor due to the neural lost and limited axonal regeneration. To date, there was no effective treatment. Recent studies have shown that bone marrow stromal cells (BMSCs) transplantated into the central nervous system (CNS) can survive and differentiate into neuronal-like cells. Additionally, granulocyte colony-stimulating factor (G-CSF) can mobilize hematopoietic stem cells and inhibit neural cell apoptosis. Thus, we aimed to evaluate the combined effect of BMSC transplantation and G-CSF administration on rats with traverse spinal cord injury. METHODS BMSCs were cultured in vitro, labeled with Hoechst33342, and then transplanted into the lesion site with or without G-CSF administration (50 microg/kg/day) for 5 subsequent days. The groups included an untreated control, along with treatment by G-CSF alone, BMSCs alone, and G-CSF + BMSCs. RESULTS In this study, by the end of eighth week after SCI injury, the animals in group treated with G-CSF + BMSCs showed higher BBB scores than the other two groups. Morphometric assessment showed that the lesion areas in the rats of the G-CSF + BMSCs group were much smaller. Compared with the control, BMSC, and G-CSF groups, less expression of apoptosis cells and more neural-cell markers around the spinal cord injury were found in rats treated with G-CSF + BMSCs. CONCLUSIONS The animals with the combination treatment achieved a better functional and morphologic recovery, although partial. This synergistic effect between BMSCs and G-CSF may be attributed to extrinsic and endogenous neurogenesis in the traverse spinal cord injury.
Collapse
|
27
|
Zhang YQ, Zeng X, He LM, Ding Y, Li Y, Zeng YS. NT-3 gene modified Schwann cells promote TrkC gene modified mesenchymal stem cells to differentiate into neuron-like cells in vitro. Anat Sci Int 2009; 85:61-7. [DOI: 10.1007/s12565-009-0056-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Accepted: 07/03/2009] [Indexed: 01/01/2023]
|
28
|
Maruichi K, Kuroda S, Chiba Y, Hokari M, Shichinohe H, Hida K, Iwasaki Y. Transplanted bone marrow stromal cells improves cognitive dysfunction due to diffuse axonal injury in rats. Neuropathology 2009; 29:422-32. [DOI: 10.1111/j.1440-1789.2008.00995.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
29
|
Askenasy N, Stein J, Farkas DL. Imaging Approaches to Hematopoietic Stem and Progenitor Cell Function and Engraftment. Immunol Invest 2009; 36:713-38. [DOI: 10.1080/08820130701715803] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
30
|
Zhu J, Zhou Z, Liu Y, Zheng J. Fractalkine and CX3CR1 are involved in the migration of intravenously grafted human bone marrow stromal cells toward ischemic brain lesion in rats. Brain Res 2009; 1287:173-83. [PMID: 19563789 DOI: 10.1016/j.brainres.2009.06.068] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2009] [Revised: 06/21/2009] [Accepted: 06/23/2009] [Indexed: 01/16/2023]
Abstract
Recent research has shown that transplanted bone marrow stromal cells (MSCs) migrate to the injured regions and exert their therapeutic effects in cases of intracranial trauma, stroke, inflammation and degenerative disease. The specific mechanisms involved in their migration to lesions are still to be fully elucidated. In the present study, a rat model of transient middle cerebral artery occlusion (MCAO) was established. At 24 h after reperfusion, human bone marrow stromal cells (hMSCs) were transplanted by intravenous injection to explore the effects of fractalkine/CX3CR1 on the migration of transplanted MSCs to lesions. In vitro study using real-time PCR and western blot revealed that CX3CR1, the only known receptor of fractalkine, was expressed in cultured hMSCs. The expression of fractalkine in the ischemic brain was significantly increased. The directional migration of transplanted hMSCs to the damaged region was observed through detection of green fluorescence protein (GFP). The results indicated the cells were mainly distributed in the ischemic boundary zone with high fractalkine expression. In a further study, lentivirus-mediated RNA interference of CX3CR1 expression was employed. The results of these experiments indicated that CX3CR1 knock-down dramatically decreased the migration of hMSCs to the ischemic brain. The present study suggests that fractalkine and its specific receptor CX3CR1 are involved in the directional migration of transplanted MSCs to the ischemic damaged brain region.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, Xinqiao Street, Chongqing 400037, PR China
| | | | | | | |
Collapse
|
31
|
Itosaka H, Kuroda S, Shichinohe H, Yasuda H, Yano S, Kamei S, Kawamura R, Hida K, Iwasaki Y. Fibrin matrix provides a suitable scaffold for bone marrow stromal cells transplanted into injured spinal cord: A novel material for CNS tissue engineering. Neuropathology 2009; 29:248-57. [DOI: 10.1111/j.1440-1789.2008.00971.x] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
32
|
Hokari M, Kuroda S, Chiba Y, Maruichi K, Iwasaki Y. Synergistic effects of granulocyte-colony stimulating factor on bone marrow stromal cell transplantation for mice cerebral infarct. Cytokine 2009; 46:260-6. [PMID: 19286390 DOI: 10.1016/j.cyto.2009.02.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2008] [Revised: 12/23/2008] [Accepted: 02/12/2009] [Indexed: 01/07/2023]
Abstract
This study was aimed to assess whether ex vivo treatment with granulocyte-colony stimulating factor (G-CSF) modifies biological properties of bone marrow stromal cells (BMSC) and enhances functional recovery by BMSC transplantation into infarct brain. Immunohistochemistry was conducted to characterize the cultured BMSC. The pharmacological effects of G-CSF on their proliferation, cell cycle, and growth factor production were precisely analyzed, using FACS and ELISA techniques. Non-treated or G-CSF treated BMSC were stereotactically transplanted into the mice brain subjected to cerebral infarct, and its effects on functional and histological aspects were evaluated. The BMSC expressed the receptor for G-CSF. Treatment with 0.1muM of G-CSF significantly enhanced the proliferation of BMSC by increasing their population in S phase, and increased their production of SDF-1alpha, HGF, and NGF. When transplanted into infarct brain, G-CSF treated BMSC significantly improved motor function as early as 2 weeks after transplantation, whereas non-treated BMSC did 4 weeks after transplantation. These findings strongly suggest that G-CSF may enhance the proliferation and growth factor production of the cultured BMSC and accelerate functional restoration by BMSC transplantation. Such pharmacological "activation" of the BMSC may contribute to successful clinical application of BMSC transplantation therapy for ischemic stroke.
Collapse
Affiliation(s)
- Masaaki Hokari
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, North 15 West 7, Kita-ku, Sapporo 060-8638, Japan
| | | | | | | | | |
Collapse
|
33
|
Chiba Y, Kuroda S, Maruichi K, Osanai T, Hokari M, Yano S, Shichinohe H, Hida K, Iwasaki Y. TRANSPLANTED BONE MARROW STROMAL CELLS PROMOTE AXONAL REGENERATION AND IMPROVE MOTOR FUNCTION IN A RAT SPINAL CORD INJURY MODEL. Neurosurgery 2009; 64:991-9; discussion 999-1000. [DOI: 10.1227/01.neu.0000341905.57162.1d] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Yasuhiro Chiba
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Satoshi Kuroda
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Katsuhiko Maruichi
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Toshiya Osanai
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masaaki Hokari
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shunsuke Yano
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hideo Shichinohe
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Kazutoshi Hida
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yoshinobu Iwasaki
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
34
|
Maruichi K, Kuroda S, Chiba Y, Hokari M, Shichinohe H, Hida K, Iwasaki Y. Graded model of diffuse axonal injury for studying head injury-induced cognitive dysfunction in rats. Neuropathology 2009; 29:132-9. [DOI: 10.1111/j.1440-1789.2008.00956.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
35
|
Hokari M, Kuroda S, Shichinohe H, Yano S, Hida K, Iwasaki Y. Bone marrow stromal cells protect and repair damaged neurons through multiple mechanisms. J Neurosci Res 2008; 86:1024-35. [DOI: 10.1002/jnr.21572] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
36
|
Kuroda S. How should we bridge the missing steps in translational research for stroke therapy?-A critical review. ACTA ACUST UNITED AC 2008. [DOI: 10.3995/jstroke.30.875] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
37
|
Deng Y, Liu Y, Zhu W, Bi X, Wang Y, Ye M, Zhou GQ. The co-transplantation of human bone marrow stromal cells and embryo olfactory ensheathing cells as a new approach to treat spinal cord injury in a rat model. Cytotherapy 2008; 10:551-64. [DOI: 10.1080/14653240802165673] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
38
|
Parr AM, Tator CH, Keating A. Bone marrow-derived mesenchymal stromal cells for the repair of central nervous system injury. Bone Marrow Transplant 2007; 40:609-19. [PMID: 17603514 DOI: 10.1038/sj.bmt.1705757] [Citation(s) in RCA: 340] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Transplantation of bone marrow-derived mesenchymal stromal cells (MSCs) into the injured brain or spinal cord may provide therapeutic benefit. Several models of central nervous system (CNS) injury have been examined, including that of ischemic stroke, traumatic brain injury and traumatic spinal cord injury in rodent, primate and, more recently, human trials. Although it has been suggested that differentiation of MSCs into cells of neural lineage may occur both in vitro and in vivo, this is unlikely to be a major factor in functional recovery after brain or spinal cord injury. Other mechanisms of recovery that may play a role include neuroprotection, creation of a favorable environment for regeneration, expression of growth factors or cytokines, vascular effects or remyelination. These mechanisms are not mutually exclusive, and it is likely that more than one contribute to functional recovery. In light of the uncertainty surrounding the fate and mechanism of action of MSCs transplanted into the CNS, further preclinical studies with appropriate animal models are urgently needed to better inform the design of new clinical trials.
Collapse
Affiliation(s)
- A M Parr
- Department of Surgery, University Health Network and Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| | | | | |
Collapse
|
39
|
Samadikuchaksaraei A. An overview of tissue engineering approaches for management of spinal cord injuries. J Neuroeng Rehabil 2007; 4:15. [PMID: 17501987 PMCID: PMC1876804 DOI: 10.1186/1743-0003-4-15] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2006] [Accepted: 05/14/2007] [Indexed: 01/09/2023] Open
Abstract
Severe spinal cord injury (SCI) leads to devastating neurological deficits and disabilities, which necessitates spending a great deal of health budget for psychological and healthcare problems of these patients and their relatives. This justifies the cost of research into the new modalities for treatment of spinal cord injuries, even in developing countries. Apart from surgical management and nerve grafting, several other approaches have been adopted for management of this condition including pharmacologic and gene therapy, cell therapy, and use of different cell-free or cell-seeded bioscaffolds. In current paper, the recent developments for therapeutic delivery of stem and non-stem cells to the site of injury, and application of cell-free and cell-seeded natural and synthetic scaffolds have been reviewed.
Collapse
Affiliation(s)
- Ali Samadikuchaksaraei
- Department of Biotechnology, Faculty of Allied Medicine and Cellular and Molecular Research Center, Iran University of Medical Sciences, Iran.
| |
Collapse
|
40
|
Yano S, Kuroda S, Shichinohe H, Seki T, Ohnishi T, Tamagami H, Hida K, Iwasaki Y. Bone marrow stromal cell transplantation preserves gammaaminobutyric acid receptor function in the injured spinal cord. J Neurotrauma 2007; 23:1682-92. [PMID: 17115913 DOI: 10.1089/neu.2006.23.1682] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A surprising shortage of information surrounds the mechanisms by which bone marrow stromal cells (BMSC) restore lost neurologic functions when transplanted into the damaged central nervous system. In the present study, we sought to elucidate whether BMSCs express the neuron-specific gamma-aminobutyric acid (GABA) receptor when transplanted into injured spinal cord. To examine this, we harvested and cultured rat femoral BMSCs. We then subjected Sprague-Dawley rats to thoracic spinal cord injury (SCI) with a pneumatic impact device. Fluorescence-labeled BMSCs (n = 7) were transplanted stereotactically or the vehicle in which these cells were cultured (n = 4) was introduced stereotactically into the rostral site of SCI at 7 days after injury. We evaluated GABA receptor function by measuring the binding potential for 125I-iomazenil (125I-IMZ) through in vitro autoradiography at 4 weeks after BMSC transplantation and simultaneously examined the fate of the transplanted BMSCs by immunocytochemistry. We found that the transplanted BMSC migrated toward the core of the injury and were densely distributed in the marginal region at 4 weeks after transplantation. BMSC transplantation significantly increased the binding potential for 125I-IMZ (p = 0.0376) and increased the number of GABA receptor-positive cells (p = 0.0077) in the marginal region of the injury site. Some of the transplanted BMSCs were positive for microtubule-associated protein-2 and the alpha1 subunit of GABA(A) receptor in the region of injury. These findings suggest that BMSCs have the potential to support the survival of neurons in the marginal region of SCI and can partly differentiate into neurons, regenerating spinal cord tissue at the site of injury.
Collapse
Affiliation(s)
- Shunsuke Yano
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
In spite of the commonly held belief that ‘the brain does not regenerate’, it is now accepted that postnatal neurogenesis does occur. Thus, one wonders whether cellular-replacement therapy might be used to heal the brain in diseases caused by neuronal cell loss. The existence of neural stem cells has been demonstrated by many scientists and is now generally accepted. The exact role of these cells, how their numbers are regulated and how they participate in CNS and spinal cord regeneration in postnatal life are still not well known. There are many reviews summarizing work on these cells; consequently, I will focus instead on other cells that may participate in postnatal neurogenesis: bone marrow-derived stem cells. The possibility that bone marrow-derived stem cells populate the CNS and differentiate into various neural elements is certainly not universally accepted.
Collapse
Affiliation(s)
- Eva Mezey
- CSDB, NIDCR, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|