1
|
Lan XY, Liang XS, Cao MX, Qin HM, Chu CY, Boltze J, Li S. NCAM mimetic peptide P2 synergizes with bone marrow mesenchymal stem cells in promoting functional recovery after stroke. J Cereb Blood Flow Metab 2024; 44:1128-1144. [PMID: 38230663 PMCID: PMC11179606 DOI: 10.1177/0271678x241226482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 11/07/2023] [Accepted: 12/08/2023] [Indexed: 01/18/2024]
Abstract
The neural cell adhesion molecule (NCAM) promotes neural development and regeneration. Whether NCAM mimetic peptides could synergize with bone marrow mesenchymal stem cells (BMSCs) in stroke treatment deserves investigation. We found that the NCAM mimetic peptide P2 promoted BMSC proliferation, migration, and neurotrophic factor expression, protected neurons from oxygen-glucose deprivation through ERK and PI3K/AKT activation and anti-apoptotic mechanisms in vitro. Following middle cerebral artery occlusion (MCAO) in rats, P2 alone or in combination with BMSCs inhibited neuronal apoptosis and induced the phosphorylation of ERK and AKT. P2 combined with BMSCs enhanced neurotrophic factor expression and BMSC proliferation in the ischemic boundary zone. Moreover, combined P2 and BMSC therapy induced translocation of nuclear factor erythroid 2-related factor, upregulated heme oxygenase-1 expression, reduced infarct volume, and increased functional recovery as compared to monotreatments. Treatment with LY294002 (PI3K inhibitor) and PD98059 (ERK inhibitor) decreased the neuroprotective effects of combined P2 and BMSC therapy in MCAO rats. Collectively, P2 is neuroprotective while P2 and BMSCs work synergistically to improve functional outcomes after ischemic stroke, which may be attributed to mechanisms involving enhanced BMSC proliferation and neurotrophic factor release, anti-apoptosis, and PI3K/AKT and ERK pathways activation.
Collapse
Affiliation(s)
- Xiao-Yan Lan
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Xue-Song Liang
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Ming-Xuan Cao
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Hua-Min Qin
- Department of Pathology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Cheng-Yan Chu
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, China
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Buchlak QD, Esmaili N, Moore J. Opportunities for developing neural stem cell treatments for acute ischemic stroke: A systematic review and gap analysis. J Clin Neurosci 2024; 120:64-75. [PMID: 38199150 DOI: 10.1016/j.jocn.2023.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Ischemic stroke is a leading cause of disability and death. Current treatments are limited. Stem cell therapy has been highlighted as a potentially effective treatment to mitigate damage and restore function, but efficacy results are mixed. This study aimed to systematically review the literature on stem cell therapies for early acute ischemic stroke; and identify opportunities for future research to facilitate the development of an effective stem cell-based treatment. Original research published within the last 10 years that focused on the evaluation of a stem cell-based treatment for acute ischemic stroke in adult patients or subjects was included. Risk of bias was assessed using the SYRCLE and Cochrane risk of bias tools for animal and human studies, respectively. 3,396 articles were screened, 58 full-text articles were reviewed and 33 met inclusion criteria. Many studies appeared to be at risk of bias. Study designs and results were heterogeneous. Most studies were preclinical and involved stem cell administration within 24 hours. Seven studies tested the effects of multiple administration timepoints and one investigated repeat dosing. Six studies were conducted in humans and stem cell administration ranged from 24 hours to 90 days post stroke. Most studies employed the use of mesenchymal stem cells. The most appropriate cell delivery method appeared to be intra-arterial. Evidence suggests that stem cell therapy may be associated with beneficial effects. A literature gap analysis identified numerous opportunities for treatment development.
Collapse
Affiliation(s)
- Quinlan D Buchlak
- Department of Neurosurgery, Monash Health, Melbourne, VIC, Australia; School of Medicine, University of Notre Dame Australia, Sydney, NSW, Australia.
| | - Nazanin Esmaili
- School of Medicine, University of Notre Dame Australia, Sydney, NSW, Australia; Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
| | - Justin Moore
- Department of Neurosurgery, Monash Health, Melbourne, VIC, Australia; Department of Surgery, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Floriano JF, Emanueli C, Vega S, Barbosa AMP, Oliveira RGD, Floriano EAF, Graeff CFDO, Abbade JF, Herculano RD, Sobrevia L, Rudge MVC. Pro-angiogenic approach for skeletal muscle regeneration. Biochim Biophys Acta Gen Subj 2022; 1866:130059. [PMID: 34793875 DOI: 10.1016/j.bbagen.2021.130059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022]
Abstract
The angiogenesis process is a phenomenon in which numerous molecules participate in the stimulation of the new vessels' formation from pre-existing vessels. Angiogenesis is a crucial step in tissue regeneration and recovery of organ and tissue function. Muscle diseases affect millions of people worldwide overcome the ability of skeletal muscle to self-repair. Pro-angiogenic therapies are key in skeletal muscle regeneration where both myogenesis and angiogenesis occur. These therapies have been based on mesenchymal stem cells (MSCs), exosomes, microRNAs (miRs) and delivery of biological factors. The use of different calls of biomaterials is another approach, including ceramics, composites, and polymers. Natural polymers are use due its bioactivity and biocompatibility in addition to its use as scaffolds and in drug delivery systems. One of these polymers is the natural rubber latex (NRL) which is biocompatible, bioactive, versatile, low-costing, and capable of promoting tissue regeneration and angiogenesis. In this review, the advances in the field of pro-angiogenic therapies are discussed.
Collapse
Affiliation(s)
- Juliana Ferreira Floriano
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; National Heart and Lung Institute, Imperial College London, London, UK.
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sofia Vega
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | | | | | | | | | - Joelcio Francisco Abbade
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil
| | | | - Luis Sobrevia
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland, Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD, 4029, Queensland, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands.
| | | |
Collapse
|
4
|
Zhang XL, Zhang XG, Huang YR, Zheng YY, Ying PJ, Zhang XJ, Lu X, Wang YJ, Zheng GQ. Stem Cell-Based Therapy for Experimental Ischemic Stroke: A Preclinical Systematic Review. Front Cell Neurosci 2021; 15:628908. [PMID: 33935650 PMCID: PMC8079818 DOI: 10.3389/fncel.2021.628908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/24/2021] [Indexed: 12/21/2022] Open
Abstract
Stem cell transplantation offers promise in the treatment of ischemic stroke. Here we utilized systematic review, meta-analysis, and meta-regression to study the biological effect of stem cell treatments in animal models of ischemic stroke. A total of 98 eligible publications were included by searching PubMed, EMBASE, and Web of Science from inception to August 1, 2020. There are about 141 comparisons, involving 5,200 animals, that examined the effect of stem cell transplantation on neurological function and infarct volume as primary outcome measures in animal models for stroke. Stem cell-based therapy can improve both neurological function (effect size, −3.37; 95% confidence interval, −3.83 to −2.90) and infarct volume (effect size, −11.37; 95% confidence interval, −12.89 to −9.85) compared with controls. These results suggest that stem cell therapy could improve neurological function deficits and infarct volume, exerting potential neuroprotective effect for experimental ischemic stroke, but further clinical studies are still needed.
Collapse
Affiliation(s)
- Xi-Le Zhang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Guang Zhang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan-Ran Huang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan-Yan Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Peng-Jie Ying
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao-Jie Zhang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiao Lu
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi-Jing Wang
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guo-Qing Zheng
- Department of Neurology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Mesenchymal stem cell therapy for ischemic stroke: A look into treatment mechanism and therapeutic potential. J Neurol 2020; 268:4095-4107. [DOI: 10.1007/s00415-020-10138-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022]
|
6
|
Qin C, Lu Y, Wang K, Bai L, Shi G, Huang Y, Li Y. Transplantation of bone marrow mesenchymal stem cells improves cognitive deficits and alleviates neuropathology in animal models of Alzheimer's disease: a meta-analytic review on potential mechanisms. Transl Neurodegener 2020; 9:20. [PMID: 32460886 PMCID: PMC7251864 DOI: 10.1186/s40035-020-00199-x] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Accepted: 05/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Alzheimer's disease is a neurodegenerative disorder. Therapeutically, a transplantation of bone marrow mesenchymal stem cells (BMMSCs) can play a beneficial role in animal models of Alzheimer's disease. However, the relevant mechanism remains to be fully elucidated. MAIN BODY Subsequent to the transplantation of BMMSCs, memory loss and cognitive impairment were significantly improved in animal models with Alzheimer's disease (AD). Potential mechanisms involved neurogenesis, apoptosis, angiogenesis, inflammation, immunomodulation, etc. The above mechanisms might play different roles at certain stages. It was revealed that the transplantation of BMMSCs could alter some gene levels. Moreover, the differential expression of representative genes was responsible for neuropathological phenotypes in Alzheimer's disease, which could be used to construct gene-specific patterns. CONCLUSIONS Multiple signal pathways involve therapeutic mechanisms by which the transplantation of BMMSCs improves cognitive and behavioral deficits in AD models. Gene expression profile can be utilized to establish statistical regression model for the evaluation of therapeutic effect. The transplantation of autologous BMMSCs maybe a prospective therapy for patients with Alzheimer's disease.
Collapse
Affiliation(s)
- Chuan Qin
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China.
| | - Yalan Lu
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Kewei Wang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Lin Bai
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Guiying Shi
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Yiying Huang
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences & Comparative Medical Center, Peking Union Medical College, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, 5 Panjiayuan Nanli St, Beijing, 100021, China
| | - Yongning Li
- Department of International Medical Service & Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shuaifuyuan 1, Dong Cheng District, Beijing, 100730, China
| |
Collapse
|
7
|
Tuazon JP, Castelli V, Borlongan CV. Drug-like delivery methods of stem cells as biologics for stroke. Expert Opin Drug Deliv 2019; 16:823-833. [PMID: 31311344 DOI: 10.1080/17425247.2019.1645116] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Stem cell therapy is an experimental treatment for brain disorders. Although a cellular product, stem cells can be classified as biologics based on the cells' secretion of therapeutic substances. Treatment with stem cell biologics may appeal to stroke because of the secondary cell death mechanisms, especially neuroinflammation, that are rampant from the onset and remain elevated during the progressive phase of the disease requiring multi-pronged biological targets to effectively abrogate the neurodegenerative pathology. However, the optimal delivery methods, among other logistical approaches (i.e. cell doses and timing of intervention), for stem cell therapy will need to be refined before stem cell biologics can be successfully utilized for stroke in large scale clinical trials. Areas covered: In this review, we discuss how the innate qualities of stem cells characterize them as biologics, how stem cell transplantation may be an ideal treatment for stroke, and the various routes of stem cell administration that have been employed in various preclinical and clinical investigations. Expert opinion: There is a need to optimize the delivery of stem cell biologics for stroke in order to guide the safe and effective translation of this therapy from the laboratory to the clinic.
Collapse
Affiliation(s)
- Julian P Tuazon
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Vanessa Castelli
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| | - Cesar V Borlongan
- a Center of Excellence for Aging & Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine , Tampa , FL , USA
| |
Collapse
|
8
|
Cui LL, Golubczyk D, Tolppanen AM, Boltze J, Jolkkonen J. Cell therapy for ischemic stroke: Are differences in preclinical and clinical study design responsible for the translational loss of efficacy? Ann Neurol 2019; 86:5-16. [PMID: 31020699 DOI: 10.1002/ana.25493] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 04/01/2019] [Accepted: 04/21/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Li-Li Cui
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| | - Dominika Golubczyk
- Department of Neurosurgery, School of Medicine, Collegium Medicum, University of Warmia and Mazury, Olsztyn, Poland
| | | | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Jukka Jolkkonen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland.,Neurocenter, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
9
|
Challenges and Controversies in Human Mesenchymal Stem Cell Therapy. Stem Cells Int 2019; 2019:9628536. [PMID: 31093291 PMCID: PMC6481040 DOI: 10.1155/2019/9628536] [Citation(s) in RCA: 325] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/12/2019] [Indexed: 12/13/2022] Open
Abstract
Stem cell therapy is being intensely investigated within the last years. Expectations are high regarding mesenchymal stem cell (MSC) treatment in translational medicine. However, many aspects concerning MSC therapy should be profoundly defined. Due to a variety of approaches that are investigated, potential effects of stem cell therapy are not transparent. On the other hand, most results of MSC administration in vivo have confirmed their safety and showed promising beneficial outcomes. However, the therapeutic effects of MSC-based treatment are still not spectacular and there is a potential risk related to MSC applications into specific cell niche that should be considered in long-term observations and follow-up outcomes. In this review, we intend to address some problems and critically discuss the complex nature of MSCs in the context of their effective and safe applications in regenerative medicine in different diseases including graft versus host disease (GvHD) and cardiac, neurological, and orthopedic disorders.
Collapse
|
10
|
Mu J, Bakreen A, Juntunen M, Korhonen P, Oinonen E, Cui L, Myllyniemi M, Zhao S, Miettinen S, Jolkkonen J. Combined Adipose Tissue-Derived Mesenchymal Stem Cell Therapy and Rehabilitation in Experimental Stroke. Front Neurol 2019; 10:235. [PMID: 30972000 PMCID: PMC6443824 DOI: 10.3389/fneur.2019.00235] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 02/22/2019] [Indexed: 01/12/2023] Open
Abstract
Background/Objective: Stroke is a leading global cause of adult disability. As the population ages as well as suffers co-morbidities, it is expected that the stroke burden will increase further. There are no established safe and effective restorative treatments to facilitate a good functional outcome in stroke patients. Cell-based therapies, which have a wide therapeutic window, might benefit a large percentage of patients, especially if combined with different restorative strategies. In this study, we tested whether the therapeutic effect of human adipose tissue-derived mesenchymal stem cells (ADMSCs) could be further enhanced by rehabilitation in an experimental model of stroke. Methods: Focal cerebral ischemia was induced in adult male Sprague Dawley rats by permanently occluding the distal middle cerebral artery (MCAO). After the intravenous infusion of vehicle (n = 46) or ADMSCs (2 × 106) either at 2 (n = 37) or 7 (n = 7) days after the operation, half of the animals were housed in an enriched environment mimicking rehabilitation. Subsequently, their behavioral recovery was assessed by a neurological score, and performance in the cylinder and sticky label tests during a 42-day behavioral follow-up. At the end of the follow-up, rats were perfused for histology to assess the extent of angiogenesis (RECA-1), gliosis (GFAP), and glial scar formation. Results: No adverse effects were observed during the follow-up. Combined ADMSC therapy and rehabilitation improved forelimb use in the cylinder test in comparison to MCAO controls on post-operative days 21 and 42 (P < 0.01). In the sticky label test, ADMSCs and rehabilitation alone or together, significantly decreased the removal time as compared to MCAO controls on post-operative days 21 and 42. An early initiation of combined therapy seemed to be more effective. Infarct size, measured by MRI on post-operative days 1 and 43, did not differ between the experimental groups. Stereological counting revealed an ischemia-induced increase both in the density of blood vessels and the numbers of glial cells in the perilesional cortex, but there were no differences among MCAO groups. Glial scar volume was also similar in MCAO groups. Conclusion: Early delivery of ADMSCs and combined rehabilitation enhanced behavioral recovery in an experimental stroke model. The mechanisms underlying these treatment effects remain unknown.
Collapse
Affiliation(s)
- Jingwei Mu
- Department of Neurology, The People's Hospital of China Medical University, Shenyang, China.,Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | | | - Miia Juntunen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Paula Korhonen
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Ella Oinonen
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Lili Cui
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Mikko Myllyniemi
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Shanshan Zhao
- Department of Neurology, University of Eastern Finland, Kuopio, Finland
| | - Susanna Miettinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Research, Development and Innovation Centre, Tampere University Hospital, Tampere, Finland
| | - Jukka Jolkkonen
- Department of Neurology, University of Eastern Finland, Kuopio, Finland.,A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.,Neurocenter, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
11
|
Xu K, Lee JY, Kaneko Y, Tuazon JP, Vale F, van Loveren H, Borlongan CV. Human stem cells transplanted into the rat stroke brain migrate to the spleen via lymphatic and inflammation pathways. Haematologica 2018; 104:1062-1073. [PMID: 30514806 PMCID: PMC6518907 DOI: 10.3324/haematol.2018.206581] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/30/2018] [Indexed: 12/13/2022] Open
Abstract
Despite mounting evidence of a massive peripheral inflammatory response accompanying stroke, the ability of intracerebrally transplanted cells to migrate to the periphery and sequester systemic inflammation remains unexamined. Here, we tested the hypothesis that human bone marrow mesenchymal stromal cells intracerebrally transplanted in the brain of adult rats subjected to experimental stroke can migrate to the spleen, a vital organ that confers peripheral inflammation after stroke. Sham or experimental stroke was induced in adult Sprague-Dawley rats by a 1 hour middle cerebral artery occlusion model. One hour after surgery, rats were intracerebrally injected with human bone marrow mesenchymal stromal cells (3×105/9 μL), then euthanized on day 1, 3, or 7 for immunohistochemical assays. Cell migration assays were performed for human bone marrow mesenchymal stromal cells using Boyden chambers with the bottom plate consisting of microglia, lymphatic endothelial cells, or both, and treated with different doses of tumor necrosis factor-α. Plates were processed in a fluorescence reader at different time points. Immunofluorescence microscopy on different days after the stroke revealed that stem cells engrafted in the stroke brain but, interestingly, homed to the spleen via lymphatic vessels, and were propelled by inflammatory signals. Experiments using human bone marrow mesenchymal stromal cells co-cultured with lymphatic endothelial cells or microglia, and treated with tumor necrosis factor-α, further indicated the key roles of the lymphatic system and inflammation in directing stem cell migration. This study is the first to demonstrate brain-to-periphery migration of stem cells, advancing the novel concept of harnessing the lymphatic system in mobilizing stem cells to sequester peripheral inflammation as a brain repair strategy.
Collapse
Affiliation(s)
- Kaya Xu
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA.,Department of Neurosurgery, The Affiliated Hospital of Guizhou Medical University, China
| | - Jea-Young Lee
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Yuji Kaneko
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Julian P Tuazon
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Fernando Vale
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Harry van Loveren
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| | - Cesario V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa, FL, USA
| |
Collapse
|
12
|
Cho DY, Jeun SS. Combination therapy of human bone marrow-derived mesenchymal stem cells and minocycline improves neuronal function in a rat middle cerebral artery occlusion model. Stem Cell Res Ther 2018; 9:309. [PMID: 30413178 PMCID: PMC6230290 DOI: 10.1186/s13287-018-1011-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/07/2018] [Accepted: 09/17/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The positive effects of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and minocycline on ischemic stroke models have been well described through numerous studies. The aim of this study was to evaluate the effectiveness of combination therapy of hBM-MSCs with minocycline in a middle cerebral artery occlusion rat model. METHODS Forty male Sprague-Dawley rats were enrolled in this study. After right middle cerebral artery occlusion, rats were randomly assigned to one of four groups: control, minocycline, hBM-MSCs, or hBM-MSCs with minocycline. Rotarod test, adhesive-removal test, and modified neurological severity score grading were performed before and 1, 7, 14, 21, and 28 days after right middle cerebral artery occlusion. All rats were sacrificed at day 28. The volume of the infarcted area was measured with triphenyl tetrazolium chloride staining. Neuronal nuclear antigen (NeuN)- and vascular endothelial growth factor (VEGF)-positive cells in the ischemic boundary zone were assessed by immunofluorescence. RESULTS Neurological outcome in the adhesive-removal test and rotarod test and modified neurological severity score were better in the combination therapy group than in the monotherapy and control groups. The volume of the infarcted area was smaller in the combination group compared with the others. The proportions of NeuN- and VEGF-positive cells in the ischemic boundary were highest in the combination therapy group. CONCLUSIONS Early combination therapy of hBM-MSCs with minocycline in an ischemic stroke model may enhance neurological recovery, reduce the volume of the infarcted area, and promote the expression of NeuN and VEGF in ischemic boundary cells.
Collapse
Affiliation(s)
- Dong Young Cho
- Department of Neurosurgery, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 137-701, Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 137-701, Korea. .,Department of Biomedical Science, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 137-701, Korea.
| |
Collapse
|
13
|
Evaluation of the Safety and Efficacy of the Therapeutic Potential of Adipose-Derived Stem Cells Injected in the Cerebral Ischemic Penumbra. J Stroke Cerebrovasc Dis 2018; 27:2453-2465. [PMID: 30029838 DOI: 10.1016/j.jstrokecerebrovasdis.2018.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 04/24/2018] [Accepted: 05/01/2018] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Stroke represents an attractive target for cell therapy. Although different types of cells have been employed in animal models with variable results, the human adipose-derived stem cells (hASCs) have demonstrated favorable characteristics in the treatment of diseases with inflammatory substrate, but experience in their intracerebral administration is lacking. The purpose of this study is to evaluate the effect and safety of the intracerebral application of hASCs in a stroke model. METHODS A first group of Athymic Nude mice after stroke received a stereotactic injection of hASCs at a concentration of 4 × 104/µL at the penumbra area, a second group without stroke received the same cell concentration, and a third group had only stroke and no cells. After 7, 15, and 30 days, the animals underwent fluorodeoxyglucose-positron emission tomography and magnetic resonance imaging; subsequently, they were sacrificed for histological evaluation (HuNu, GFAP, IBA-1, Ki67, DCX) of the penumbra area and ipsilateral subventricular zone (iSVZ). RESULTS The in vitro studies found no alterations in the molecular karyotype, clonogenic capacity, and expression of 62 kDa transcription factor and telomerase. Animals implanted with cells showed no adverse events. The implanted cells showed no evidence of proliferation or differentiation. However, there was an increase of capillaries, less astrocytes and microglia, and increased bromodeoxyuridine and doublecortin-positive cells in the iSVZ and in the vicinity of ischemic injury. CONCLUSIONS These results suggest that hASCs in the implanted dose modulate inflammation, promote endogenous neurogenesis, and do not proliferate or migrate in the brain. These data confirm the safety of cell therapy with hASCs.
Collapse
|
14
|
Yamaguchi S, Horie N, Satoh K, Ishikawa T, Mori T, Maeda H, Fukuda Y, Ishizaka S, Hiu T, Morofuji Y, Izumo T, Nishida N, Matsuo T. Age of donor of human mesenchymal stem cells affects structural and functional recovery after cell therapy following ischaemic stroke. J Cereb Blood Flow Metab 2018; 38:1199-1212. [PMID: 28914133 PMCID: PMC6434451 DOI: 10.1177/0271678x17731964] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cell transplantation therapy offers great potential to improve impairments after stroke. However, the importance of donor age on therapeutic efficacy is unclear. We investigated the regenerative capacity of transplanted cells focusing on donor age (young vs. old) for ischaemic stroke. The quantities of human mesenchymal stem cell (hMSC) secreted brain-derived neurotrophic factor in vitro and of monocyte chemotactic protein-1 at day 7 in vivo were both significantly higher for young hMSC compared with old hMSC. Male Sprague-Dawley rats subjected to transient middle cerebral artery occlusion that received young hMSC (trans-arterially at 24 h after stroke) showed better behavioural recovery with prevention of brain atrophy compared with rats that received old hMSC. Histological analysis of the peri-infarct cortex showed that rats treated with young hMSC had significantly fewer microglia and more vessels covered with pericytes. Interestingly, migration of neural stem/progenitor cells expressing Musashi-1 positively correlated with astrocyte process alignment, which was more pronounced for young hMSC. Aging of hMSC may be a critical factor that affects cell therapy outcomes, and transplantation of young hMSC appears to provide better functional recovery through anti-inflammatory effects, vessel maturation, and neurogenesis potentially by the dominance of trophic factor secretion.
Collapse
Affiliation(s)
- Susumu Yamaguchi
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Nobutaka Horie
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Katsuya Satoh
- 2 Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takeshi Ishikawa
- 2 Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tsuyoshi Mori
- 2 Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Hajime Maeda
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yuhtaka Fukuda
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shunsuke Ishizaka
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takeshi Hiu
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yoichi Morofuji
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tsuyoshi Izumo
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Noriyuki Nishida
- 2 Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takayuki Matsuo
- 1 Department of Neurosurgery, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
15
|
Zheng Z, Zhang L, Qu Y, Xiao G, Li S, Bao S, Lu QR, Mu D. Mesenchymal Stem Cells Protect Against Hypoxia-Ischemia Brain Damage by Enhancing Autophagy Through Brain Derived Neurotrophic Factor/Mammalin Target of Rapamycin Signaling Pathway. Stem Cells 2018; 36:1109-1121. [PMID: 29451335 DOI: 10.1002/stem.2808] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/06/2018] [Accepted: 02/10/2018] [Indexed: 12/17/2022]
Abstract
Hypoxic-ischemic encephalopathy (HIE) is a serious disease for neonates. However, present therapeutic strategies are not effective enough for treating HIE. Previous study showed that mesenchymal stem cells (MSCs) can exert neuroprotective effects for brain damage, but its mechanism remains elusive. Using in vitro coculture of rat cortical primary neurons and MSCs in HI conditions, we demonstrated that MSCs help increase brain derived neurotrophic factor (BDNF) and autophagy markers (LC3II and Beclin1) in the cultures and decrease cells death (lactate dehydrogenase levels). We demonstrated a similar mechanism using an in vivo rat model of HI in combination with MSCs transplantation. Using a behavioral study, we further showed that MSCs transplantation into the rat brain after HI injury can attenuate behavioral deficits. Finally, we found that the increase in BDNF and autophagy related factors after HI injury combined with MSCs transplantation can be reversed by anti-BDNF treatment and strengthen the point that the protective effects of BDNF work through inhibition of the mammalin target of rapamycin (mTOR) pathway. Collectively, we proposed that coculture/transplantation of MSCs after HI injury leads to increased BDNF expression and a subsequent reduction in mTOR pathway activation that results in increased autophagy and neuroprotection. This finding gives a hint to explore new strategies for treating neonates with HIE. Stem Cells 2018;36:1109-1121.
Collapse
Affiliation(s)
- Zhen Zheng
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China.,Department of Pediatrics, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Li Zhang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Guoguang Xiao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Shiping Li
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Shan Bao
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Q Richard Lu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, People's Republic of China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
16
|
Zhao LR, Willing A. Enhancing endogenous capacity to repair a stroke-damaged brain: An evolving field for stroke research. Prog Neurobiol 2018; 163-164:5-26. [PMID: 29476785 PMCID: PMC6075953 DOI: 10.1016/j.pneurobio.2018.01.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 01/11/2018] [Accepted: 01/30/2018] [Indexed: 02/07/2023]
Abstract
Stroke represents a severe medical condition that causes stroke survivors to suffer from long-term and even lifelong disability. Over the past several decades, a vast majority of stroke research targets neuroprotection in the acute phase, while little work has been done to enhance stroke recovery at the later stage. Through reviewing current understanding of brain plasticity, stroke pathology, and emerging preclinical and clinical restorative approaches, this review aims to provide new insights to advance the research field for stroke recovery. Lifelong brain plasticity offers the long-lasting possibility to repair a stroke-damaged brain. Stroke impairs the structural and functional integrity of entire brain networks; the restorative approaches containing multi-components have great potential to maximize stroke recovery by rebuilding and normalizing the stroke-disrupted entire brain networks and brain functioning. The restorative window for stroke recovery is much longer than previously thought. The optimal time for brain repair appears to be at later stage of stroke rather than the earlier stage. It is expected that these new insights will advance our understanding of stroke recovery and assist in developing the next generation of restorative approaches for enhancing brain repair after stroke.
Collapse
Affiliation(s)
- Li-Ru Zhao
- Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Alison Willing
- Center for Excellence in Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida, Morsani College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
17
|
Zhang K, Shi Z, Zhou J, Xing Q, Ma S, Li Q, Zhang Y, Yao M, Wang X, Li Q, Li J, Guan F. Potential application of an injectable hydrogel scaffold loaded with mesenchymal stem cells for treating traumatic brain injury. J Mater Chem B 2018; 6:2982-2992. [DOI: 10.1039/c7tb03213g] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this contribution, we developed an injectable hydrogel composed of sodium alginate and hyaluronic acid that acts as a tissue scaffold to create a more optimal microenvironment for the stem cells for potential application of traumatic brain injury implantation.
Collapse
|
18
|
Smith HK, Omura S, Vital SA, Becker F, Senchenkova EY, Kaur G, Tsunoda I, Peirce SM, Gavins FNE. Metallothionein I as a direct link between therapeutic hematopoietic stem/progenitor cells and cerebral protection in stroke. FASEB J 2017; 32:2381-2394. [PMID: 29269399 DOI: 10.1096/fj.201700746r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Stroke continues to be a leading cause of death and disability worldwide, yet effective treatments are lacking. Previous studies have indicated that stem-cell transplantation could be an effective treatment. However, little is known about the direct impact of transplanted cells on injured brain tissue. We wanted to help fill this knowledge gap and investigated effects of hematopoietic stem/progenitor cells (HSPCs) on the cerebral microcirculation after ischemia-reperfusion injury (I/RI). Treatment of HSPCs in I/RI for up to 2 wk after cerebral I/RI led to decreased mortality rate, decreased infarct volume, improved functional outcome, reduced microglial activation, and reduced cerebral leukocyte adhesion. Confocal microscopy and fluorescence-activated cell sorting analyses showed transplanted HSPCs emigrate preferentially into ischemic cortex brain parenchyma. We isolated migrated HSPCs from the brain; using RNA sequencing to investigate the transcriptome, we found metallothionein (MT, particularly MT-I) transcripts were dramatically up-regulated. Finally, to confirm the significance of MT, we exogenously administered MT-I after cerebral I/RI and found that it produced neuroprotection in a manner similar to HSPC treatment. These findings provide novel evidence that the mechanism through which HSPCs promote repair after stroke maybe via direct action of HSPC-derived MT-I and could therefore be exploited as a useful therapeutic strategy for stroke.-Smith, H. K., Omura, S., Vital, S. A., Becker, F., Senchenkova, E. Y., Kaur, G., Tsunoda, I., Peirce, S. M., Gavins, F. N. E. Metallothionein I as a direct link between therapeutic hematopoietic stem/progenitor cells and cerebral protection in stroke.
Collapse
Affiliation(s)
- Helen K Smith
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA.,Pathology and Laboratory Medicine, Weill Cornell Medical College, New York City, New York, USA
| | - Seiichi Omura
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA.,Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan
| | - Shantel A Vital
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| | - Felix Becker
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA.,Department for General and Visceral Surgery, University Hospital Muenster, Muenster, Germany
| | - Elena Y Senchenkova
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| | - Gaganpreet Kaur
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| | - Ikuo Tsunoda
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA.,Department of Microbiology, Kindai University Faculty of Medicine, Osakasayama, Osaka, Japan.,Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Felicity N E Gavins
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA.,Department of Neurology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
19
|
Sarmah D, Agrawal V, Rane P, Bhute S, Watanabe M, Kalia K, Ghosh Z, Dave KR, Yavagal DR, Bhattacharya P. Mesenchymal Stem Cell Therapy in Ischemic Stroke: A Meta-analysis of Preclinical Studies. Clin Pharmacol Ther 2017; 103:990-998. [PMID: 29090465 DOI: 10.1002/cpt.927] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/17/2017] [Accepted: 10/18/2017] [Indexed: 12/16/2022]
Abstract
Numerous preclinical studies have been carried out using mesenchymal stem cells (MSCs) therapy for ischemic stroke. The purpose of the present meta-analysis is to review the quality of preclinical studies. In all, 4,361 articles were identified, out of which 64 studies were included (excluding in vitro studies). The results were obtained across species, route, and time of administration, immunogenicity, and doses. The median quality score 4.90/10, confidence interval 95%, and large effect size were observed, which strongly supports the translation potential of MSC therapy for ischemic stroke.
Collapse
Affiliation(s)
- Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Vishal Agrawal
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Pallavi Rane
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Shashikala Bhute
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Mitsuyoshi Watanabe
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
| | - Zhumur Ghosh
- Department of Bioinformatics, Bose Institute, Kolkata, India
| | - Kunjan R Dave
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Dileep R Yavagal
- Department of Neurology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat, India
- Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Doeppner TR, Bähr M, Hermann DM, Giebel B. Concise Review: Extracellular Vesicles Overcoming Limitations of Cell Therapies in Ischemic Stroke. Stem Cells Transl Med 2017; 6:2044-2052. [PMID: 28941317 PMCID: PMC6430061 DOI: 10.1002/sctm.17-0081] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/05/2017] [Indexed: 12/11/2022] Open
Abstract
Despite recent advances in stroke therapy, current therapeutic concepts are still limited. Thus, additional therapeutic strategies are in order. In this sense, the transplantation of stem cells has appeared to be an attractive adjuvant tool to help boost the endogenous regenerative capacities of the brain. Although transplantation of stem cells is known to induce beneficial outcome in (preclinical) stroke research, grafted cells do not replace lost tissue directly. Rather, these transplanted cells like neural progenitor cells or mesenchymal stem cells act in an indirect manner, among which the secretion of extracellular vesicles (EVs) appears to be one key factor. Indeed, the application of EVs in preclinical stroke studies suggests a therapeutic role, which appears to be noninferior in comparison to the transplantation of stem cells themselves. In this short review, we highlight some of the recent advances in the field of EVs as a therapeutic means to counter stroke. Stem Cells Translational Medicine2017;6:2044–2052
Collapse
Affiliation(s)
- Thorsten R Doeppner
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Mathias Bähr
- Department of Neurology, University Medical Center Goettingen, Goettingen, Germany
| | - Dirk M Hermann
- Department of Neurology, University of Duisburg-Essen Medical School, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University of Duisburg-Essen Medical School, Essen, Germany
| |
Collapse
|
21
|
Zheng WP, Zhang BY, Shen ZY, Yin ML, Cao Y, Song HL. Biological effects of bone marrow mesenchymal stem cells on hepatitis B virus in vitro. Mol Med Rep 2017; 15:2551-2559. [PMID: 28447750 PMCID: PMC5428401 DOI: 10.3892/mmr.2017.6330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 12/09/2016] [Indexed: 02/07/2023] Open
Abstract
The aim of the present study was to explore the effects of co‑culturing bone marrow‑derived mesenchymal stem cells (BM-MSCs) cultured with hepatitis B virus (HBV)‑infected lymphocytes in vitro. BM‑MSCs and lymphocytes from Brown Norway rats were obtained from the bone marrow and spleen, respectively. Rats were divided into the following five experimental groups: Group 1, splenic lymphocytes (SLCs); group 2, HepG2.2.15 cells; group 3, BM‑MSCs + HepG2.2.15 cells; group 4, SLCs + HepG2.2.15 cells; and group 5, SLCs + BM‑MSCs + HepG2.2.15 cells. The viability of lymphocytes and HepG2.2.15 cells was assessed using the MTT assay at 24, 48 and 72 h, respectively. Levels of supernatant HBV DNA and intracellular HBV covalently closed circular DNA (cccDNA) were measured using quantitative polymerase chain reaction. Supernatant cytokine levels were measured by enzyme‑linked immunosorbent assay (ELISA). T cell subsets were quantified by flow cytometry using fluorescence‑labeled antibodies. In addition, the HBV genome sequence was analyzed by direct gene sequencing. Levels of HBV DNA and cccDNA in group 5 were lower when compared with those in group 3 or group 4, with a significant difference observed at 48 h. The secretion of interferon‑γ was negatively correlated with the level of HBV DNA, whereas secretion of interleukin (IL)‑10 and IL‑22 were positively correlated with the level of HBV DNA. Flow cytometry demonstrated that the percentage of CD3+CD8+ T cells was positively correlated with the levels of HBV DNA, and the CD3+CD4+/CD3+CD8+ ratio was negatively correlated with the level of HBV DNA. Almost no mutations in the HBV DNA sequence were detected in HepG2.2.15 cells co‑cultured with BM‑MSCs, SLCs, or in the two types of cells combined. BM‑MSCs inhibited the expression of HBV DNA and enhanced the clearance of HBV, which may have been mediated by the regulation of the Tc1/Tc2 cell balance and the mode of cytokine secretion to modulate cytokine expression.
Collapse
Affiliation(s)
- Wei-Ping Zheng
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Bo-Ya Zhang
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Zhong-Yang Shen
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
| | - Ming-Li Yin
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Yi Cao
- Tianjin First Central Hospital Clinic Institute, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Hong-Li Song
- Department of Organ Transplantation, Tianjin First Central Hospital, Tianjin 300192, P.R. China
- Tianjin Key Laboratory of Organ Transplantation, Tianjin 300192, P.R. China
| |
Collapse
|
22
|
Nitzsche F, Müller C, Lukomska B, Jolkkonen J, Deten A, Boltze J. Concise Review: MSC Adhesion Cascade-Insights into Homing and Transendothelial Migration. Stem Cells 2017; 35:1446-1460. [DOI: 10.1002/stem.2614] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/13/2017] [Accepted: 02/23/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Franziska Nitzsche
- Department of Ischemia Research; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Department of Radiology, McGowan Institute for Regenerative Medicine; University of Pittsburgh; Pittsburgh Pennsylvania USA
| | - Claudia Müller
- Department of Ischemia Research; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
| | - Barbara Lukomska
- NeuroRepair Department; Mossakowski Medical Research Centre; Warsaw Poland
| | - Jukka Jolkkonen
- Department of Neurology; Institute of Clinical Medicine, University of Eastern; Kuopio Finland
| | - Alexander Deten
- Translational Centre for Regenerative Medicine, Leipzig University; Leipzig Germany
| | - Johannes Boltze
- Department of Ischemia Research; Fraunhofer Institute for Cell Therapy and Immunology; Leipzig Germany
- Translational Centre for Regenerative Medicine, Leipzig University; Leipzig Germany
- Department of Translational Medicine and Cell Technology; Fraunhofer Research Institution for Marine Biotechnology and Institute for Medical and Marine Biotechnology, University of Lübeck; Lübeck Germany
| |
Collapse
|
23
|
Choi YK, Urnukhsaikhan E, Yoon HH, Seo YK, Park JK. Effect of human mesenchymal stem cell transplantation on cerebral ischemic volume-controlled photothrombotic mouse model. Biotechnol J 2016; 11:1397-1404. [PMID: 27440447 PMCID: PMC5132146 DOI: 10.1002/biot.201600057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/12/2016] [Accepted: 07/14/2016] [Indexed: 12/03/2022]
Abstract
Various animal models of stroke have been developed to simulate the human stroke with the development of the ischemic method facilitates preclinical stroke research. The photothrombotic ischemia model, based on the intravascular photochemical reaction, is widely used for in vivo studies. However, this study has limitations, which generated a relatively small‐sized infarction model on superficial cortex compared to that of the MCAO stroke model. In this study, the photothorombosis mouse model is adapted and the optimum conditions for generation of cell death and deficits with high reproducibility is determined. The extent of damage within the cortex was assessed by infarct volume and cellular/behavioral analyses. In this model, the neural cell death and inflammatory responses is detected; moreover, the degree of behavioral impairment is correlated with the brain infarct volume. Further, to enhance the understanding of neural repair, the effect of neural differentiation by transplantation of human bone marrow‐derived mesenchymal stem cells (BM‐MSCs) is analyzed. The authors demonstrated that transplantation of BM‐MSCs promoted the neural differentiation and behavioral performance in their photothrombosis model. Therefore, this research was meaningful to provide a stable animal model of stroke with low variability. Moreover, this model will facilitate development of novel MSC‐based therapeutics for stroke.
Collapse
Affiliation(s)
- Yun-Kyong Choi
- Department of Medical Biotechnology, Dongguk University, Gyeonggi-do, Republic of Korea
| | - Enerelt Urnukhsaikhan
- Department of Medical Biotechnology, Dongguk University, Gyeonggi-do, Republic of Korea
| | - Hee-Hoon Yoon
- Department of Medical Biotechnology, Dongguk University, Gyeonggi-do, Republic of Korea
| | - Young-Kwon Seo
- Department of Medical Biotechnology, Dongguk University, Gyeonggi-do, Republic of Korea
| | - Jung-Keug Park
- Department of Medical Biotechnology, Dongguk University, Gyeonggi-do, Republic of Korea
| |
Collapse
|
24
|
Li G, Yu F, Lei T, Gao H, Li P, Sun Y, Huang H, Mu Q. Bone marrow mesenchymal stem cell therapy in ischemic stroke: mechanisms of action and treatment optimization strategies. Neural Regen Res 2016; 11:1015-24. [PMID: 27482235 PMCID: PMC4962565 DOI: 10.4103/1673-5374.184506] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Animal and clinical studies have confirmed the therapeutic effect of bone marrow mesenchymal stem cells on cerebral ischemia, but their mechanisms of action remain poorly understood. Here, we summarize the transplantation approaches, directional migration, differentiation, replacement, neural circuit reconstruction, angiogenesis, neurotrophic factor secretion, apoptosis, immunomodulation, multiple mechanisms of action, and optimization strategies for bone marrow mesenchymal stem cells in the treatment of ischemic stroke. We also explore the safety of bone marrow mesenchymal stem cell transplantation and conclude that bone marrow mesenchymal stem cell transplantation is an important direction for future treatment of cerebral ischemia. Determining the optimal timing and dose for the transplantation are important directions for future research.
Collapse
Affiliation(s)
- Guihong Li
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China; Department of Neurosurgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Fengbo Yu
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| | - Ting Lei
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Haijun Gao
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Peiwen Li
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yuxue Sun
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Haiyan Huang
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Qingchun Mu
- Department of Neurosurgery, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, Heilongjiang Province, China
| |
Collapse
|
25
|
Rodríguez-Frutos B, Otero-Ortega L, Gutiérrez-Fernández M, Fuentes B, Ramos-Cejudo J, Díez-Tejedor E. Stem Cell Therapy and Administration Routes After Stroke. Transl Stroke Res 2016; 7:378-87. [PMID: 27384771 DOI: 10.1007/s12975-016-0482-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 06/21/2016] [Accepted: 06/27/2016] [Indexed: 12/23/2022]
Abstract
Cell-based therapy has demonstrated safety and efficacy in experimental animal models of stroke, as well as safety in stroke patients. However, various questions remain regarding the therapeutic window, dosage, route of administration, and the most appropriate cell type and source, as well as mechanisms of action and immune-modulation to optimize treatment based on stem cell therapy. Various delivery routes have been used in experimental stroke models, including intracerebral, intraventricular, subarachnoid, intra-arterial, intraperitoneal, intravenous, and intranasal routes. From a clinical point of view, it is necessary to demonstrate which is the most feasible, safest, and most effective for use with stroke patients. Therefore, further experimental studies concerning the safety, efficacy, and mechanisms of action involved in these therapeutic effects are required to determine their optimal clinical use.
Collapse
Affiliation(s)
- Berta Rodríguez-Frutos
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, Neuroscience Area of IdiPAZ (Health Research Institute), Autonomous University of Madrid, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Laura Otero-Ortega
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, Neuroscience Area of IdiPAZ (Health Research Institute), Autonomous University of Madrid, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - María Gutiérrez-Fernández
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, Neuroscience Area of IdiPAZ (Health Research Institute), Autonomous University of Madrid, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain.
| | - Blanca Fuentes
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, Neuroscience Area of IdiPAZ (Health Research Institute), Autonomous University of Madrid, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Jaime Ramos-Cejudo
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, Neuroscience Area of IdiPAZ (Health Research Institute), Autonomous University of Madrid, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Exuperio Díez-Tejedor
- Department of Neurology and Stroke Center, Neuroscience and Cerebrovascular Research Laboratory, Neuroscience Area of IdiPAZ (Health Research Institute), Autonomous University of Madrid, La Paz University Hospital, Paseo de la Castellana 261, 28046, Madrid, Spain.
| |
Collapse
|
26
|
Jolkkonen J, Kwakkel G. Translational Hurdles in Stroke Recovery Studies. Transl Stroke Res 2016; 7:331-42. [PMID: 27000881 DOI: 10.1007/s12975-016-0461-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/11/2016] [Accepted: 03/09/2016] [Indexed: 12/20/2022]
Abstract
Emerging understanding of brain plasticity has opened new avenues for the treatment of stroke. The promising preclinical evidence with neuroprotective drugs has not been confirmed in clinical trials, thus nowadays, researchers, pharmaceutical companies, and funding bodies hesitate to initiate these expensive trials with restorative therapies. Since many of the previous failures can be traced to low study quality, a number of guidelines such as STAIR and STEPS were introduced to rectify these shortcomings. However, these guidelines stem from the study design for neuroprotective drugs and one may question whether they are appropriate for restorative approaches, which rely heavily on behavioral testing. Most of the recovery studies conducted in stroke patients have been small-scale, proof-of-concept trials. Consequently, the overall effect sizes of pooled phase II trials have proved unreliable and unstable in most meta-analyses. Although the methodological quality of trials in humans is improving, most studies still suffer from methodological flaws and do not meet even the minimum of evidence-based standards for reporting randomized controlled trials. The power problem of most phase II trials is mostly attributable to a lack of proper stratification with robust prognostic factors at baseline as well as the incorrect assumption that all patients will exhibit the same proportional amount of spontaneous neurological recovery poststroke. In addition, most trials suffer from insufficient treatment contrasts between the experimental and control arm and the outcomes have not been sufficiently responsive to detect small but clinically relevant changes in neurological impairments and activities. This narrative review describes the main factors that bias recovery studies, both in experimental animals and stroke patients.
Collapse
Affiliation(s)
- Jukka Jolkkonen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, Kuopio, Finland. .,Neurocenter, Neurology, University Hospital of Kuopio, Kuopio, Finland.
| | - Gert Kwakkel
- Department of Rehabilitation Medicine, VU University Medical Center, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands.,Neurorehabilitation, Amsterdam Rehabilitation Research Center, Reade, Amsterdam, The Netherlands.,Neuroscience Campus Amsterdam, VU University Amsterdam, Amsterdam, The Netherlands.,Department of Physical Therapy and Human Movement Sciences, Northwestern University, Evanston, IL, USA
| |
Collapse
|
27
|
Moisan A, Favre I, Rome C, De Fraipont F, Grillon E, Coquery N, Mathieu H, Mayan V, Naegele B, Hommel M, Richard MJ, Barbier EL, Remy C, Detante O. Intravenous Injection of Clinical Grade Human MSCs After Experimental Stroke: Functional Benefit and Microvascular Effect. Cell Transplant 2016; 25:2157-2171. [PMID: 26924704 DOI: 10.3727/096368916x691132] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Stroke is the leading cause of disability in adults. Many current clinical trials use intravenous (IV) administration of human bone marrow-derived mesenchymal stem cells (BM-MSCs). This autologous graft requires a delay for ex vivo expansion of cells. We followed microvascular effects and mechanisms of action involved after an IV injection of human BM-MSCs (hBM-MSCs) at a subacute phase of stroke. Rats underwent a transient middle cerebral artery occlusion (MCAo) or a surgery without occlusion (sham) at day 0 (D0). At D8, rats received an IV injection of 3 million hBM-MSCs or PBS-glutamine. In a longitudinal behavioral follow-up, we showed delayed somatosensory and cognitive benefits 4 to 7 weeks after hBM-MSC injection. In a separate longitudinal in vivo magnetic resonance imaging (MRI) study, we observed an enhanced vascular density in the ischemic area 2 and 3 weeks after hBM-MSC injection. Histology and quantitative polymerase chain reaction (qPCR) revealed an overexpression of angiogenic factors such as Ang1 and transforming growth factor-1 (TGF-1) at D16 in hBM-MSC-treated MCAo rats compared to PBS-treated MCAo rats. Altogether, delayed IV injection of hBM-MSCs provides functional benefits and increases cerebral angiogenesis in the stroke lesion via a release of endogenous angiogenic factors enhancing the stabilization of newborn vessels. Enhanced angiogenesis could therefore be a means of improving functional recovery after stroke.
Collapse
|
28
|
Jendelová P, Kubinová Š, Sandvig I, Erceg S, Sandvig A, Syková E. Current developments in cell- and biomaterial-based approaches for stroke repair. Expert Opin Biol Ther 2015; 16:43-56. [DOI: 10.1517/14712598.2016.1094457] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
29
|
Acosta SA, Tajiri N, Hoover J, Kaneko Y, Borlongan CV. Intravenous Bone Marrow Stem Cell Grafts Preferentially Migrate to Spleen and Abrogate Chronic Inflammation in Stroke. Stroke 2015. [PMID: 26219646 PMCID: PMC4542567 DOI: 10.1161/strokeaha.115.009854] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND PURPOSE Adult stem cell therapy is an experimental stroke treatment. Here, we assessed homing and anti-inflammatory effects of bone marrow stromal cells (hBMSCs) in chronic stroke. METHODS At 60 days post stroke, adult Sprague-Dawley rats received intravenous hBMSCs (4×10(6) labeled or nonlabeled cells) or vehicle (saline). A sham surgery group served as additional control. In vivo imaging was conducted between 1 hour and 11 days post transplantation, followed by histological examination. RESULTS Labeled hBMSCs migrated to spleen which emitted significantly higher fluorescent signal across all time points, especially during the first hour, and were modestly detected in the head region at the 12 hours and 11 days, compared with nonlabeled hBMSCs and vehicle-infused stroke animals, or sham (P<0.05). At 11 days post transplantation, ex vivo imaging confirmed preferential hBMSC migration to the spleen over the brain. Hematoxylin and eosin staining revealed significant 15% and 30% reductions in striatal infarct and peri-infarct area, and a trend of rescue against neuronal loss in the hippocampus. Unbiased stereology showed significant 75% and 60% decrements in major histocompatibility complex II-activated inflammatory cells in gray and white matter, and a 43% diminution in tumor necrosis factor-α cell density in the spleen of transplanted stroke animals compared with vehicle-infused stroke animals (P<0.05). Human antigen immunostaining revealed 0.03% hBMSCs survived in spleen and only 0.0007% in brain. MSC migration to spleen, but not brain, inversely correlated with reduced infarct, peri-infarct, and inflammation. CONCLUSIONS hBMSC transplantation is therapeutic in chronic stroke possibly by abrogating the inflammation-plagued secondary cell death.
Collapse
Affiliation(s)
- Sandra A Acosta
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa
| | - Naoki Tajiri
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa
| | - Jaclyn Hoover
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa
| | - Yuji Kaneko
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa
| | - Cesar V Borlongan
- From the Center of Excellence for Aging and Brain Repair, Department of Neurosurgery and Brain Repair, University of South Florida College of Medicine, Tampa.
| |
Collapse
|
30
|
Cai Q, Chen Z, Kong DK, Wang J, Xu Z, Liu B, Chen Q, Zhou J. Novel microcatheter-based intracarotid delivery approach for MCAO/R mice. Neurosci Lett 2015; 597:127-31. [PMID: 25899778 DOI: 10.1016/j.neulet.2015.04.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/15/2015] [Accepted: 04/17/2015] [Indexed: 10/23/2022]
Abstract
The intra-arterial (IA) model by microcatheter administration was an effective way to deliver drugs or cells to the brain. All of these models were carried out introduced in rat rather than mice for the difficult and technically challenging due to their small caliber. In 2014, Alejandro Santillan first introduced this model in mice and we found that most of the operational steps were similar with the middle cerebral artery occlusion and reperfusion (MCAO/R) model. We attempted to combine these two techniques into a single model in mice and discovered that this technique was indeed possible. In our work, 12C57Bl/6J male mice were carried on middle cerebral artery occlusion for 60min and then the intra-arterial microcatheter was placed into the internal carotid artery (ICA) from the external carotid artery (ECA). GFP-Luc-Pro labeled mNSCs were infused through the microcatheter and then the blood flow perfusion was reestablished subsequently. The results showed that all 12 mice were carried on successfully the model of middle cerebral artery occlusion, and the placement of the microcatheter and the mNSCs perfusion were completed smoothly without exception. Which means that it is logical to combine the two models into one in order to facilitate studying of stroke. Meanwhile, during the dissection, we found the variation of occipital artery (OA) was noticeable and we classified first time this variation into four categories to attempt to protect the OA.
Collapse
Affiliation(s)
- Qiang Cai
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, Hubei Province, 430060, China; Department of Neurosurgery, Yale University, 333 Cedar Street, FMB 410, New Haven, CT 06520, USA
| | - Zhibiao Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, Hubei Province, 430060, China
| | - Derek Kai Kong
- Department of Neurosurgery, Yale University, 333 Cedar Street, FMB 410, New Haven, CT 06520, USA
| | - Jun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, Hubei Province, 430060, China
| | - Zhou Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, Hubei Province, 430060, China
| | - Baohui Liu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, Hubei Province, 430060, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan City, Hubei Province, 430060, China.
| | - Jiangbing Zhou
- Department of Neurosurgery, Yale University, 333 Cedar Street, FMB 410, New Haven, CT 06520, USA
| |
Collapse
|
31
|
Fukuda Y, Horie N, Satoh K, Yamaguchi S, Morofuji Y, Hiu T, Izumo T, Hayashi K, Nishida N, Nagata I. Intra-arterial transplantation of low-dose stem cells provides functional recovery without adverse effects after stroke. Cell Mol Neurobiol 2015; 35:399-406. [PMID: 25398358 DOI: 10.1007/s10571-014-0135-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 10/29/2014] [Indexed: 11/28/2022]
Abstract
Cell transplantation therapy for cerebral infarction has emerged as a promising treatment to reduce brain damage and enhance functional recovery. We previously reported that intra-arterial delivery of bone marrow mesenchymal stem cells (MSCs) enables superselective cell administration to the infarct area and results in significant functional recovery after ischemic stroke in a rat model. However, to reduce the risk of embolism caused by the transplanted cells, an optimal cell number should be determined. At 24 h after middle cerebral artery occlusion and reperfusion, we administered human MSCs (low dose: 1 × 10(4) cells; high dose: 1 × 10(6) cells) and then assessed functional recovery, inflammatory responses, cell distribution, and mortality. Rats treated with high- or low-dose MSCs showed behavioral recovery. At day 8 post-stroke, microglial activation was suppressed significantly, and interleukin (IL)-1β and IL-12p70 were reduced in both groups. Although high-dose MSCs were more widely distributed in the cortex and striatum of rats, the degree of intravascular cell aggregation and mortality was significantly higher in the high-dose group. In conclusion, selective intra-arterial transplantation of low-dose MSCs has anti-inflammatory effects and reduces the adverse effects of embolic complication, resulting in sufficient functional recovery of the affected brain.
Collapse
Affiliation(s)
- Yuhtaka Fukuda
- Department of Neurosurgery, School of Medicine, Nagasaki University, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Nobutaka Horie
- Department of Neurosurgery, School of Medicine, Nagasaki University, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan.
| | - Katsuya Satoh
- Department of Molecular Microbiology and Immunology, School of Medicine, Nagasaki University, Nagasaki, Japan
| | - Susumu Yamaguchi
- Department of Neurosurgery, School of Medicine, Nagasaki University, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Youichi Morofuji
- Department of Neurosurgery, School of Medicine, Nagasaki University, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Takeshi Hiu
- Department of Neurosurgery, School of Medicine, Nagasaki University, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Tsuyoshi Izumo
- Department of Neurosurgery, School of Medicine, Nagasaki University, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Kentaro Hayashi
- Department of Neurosurgery, School of Medicine, Nagasaki University, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| | - Noriyuki Nishida
- Department of Molecular Microbiology and Immunology, School of Medicine, Nagasaki University, Nagasaki, Japan
| | - Izumi Nagata
- Department of Neurosurgery, School of Medicine, Nagasaki University, 1-7-1, Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
32
|
Dulamea AO. The potential use of mesenchymal stem cells in stroke therapy--From bench to bedside. J Neurol Sci 2015; 352:1-11. [PMID: 25818674 DOI: 10.1016/j.jns.2015.03.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/09/2015] [Accepted: 03/10/2015] [Indexed: 12/11/2022]
Abstract
Stroke is the second main cause of morbidity and mortality worldwide. The rationale for the use of mesenchymal stem cells (MSCs) in stroke is based on the capacity of MSCs to secrete a large variety of bioactive molecules such as growth factors, cytokines and chemokines leading to reduction of inflammation, increased neurogenesis from the germinative niches of central nervous system, increased angiogenesis, effects on astrocytes, oligodendrocytes and axons. This review presents the data derived from experimental studies and the evidence available from clinical trials about the use of MSCs in stroke therapy.
Collapse
Affiliation(s)
- Adriana Octaviana Dulamea
- U.M.F. "Carol Davila", Fundeni Clinical Institute, Department of Neurology, 258 Sos. Fundeni, Sector 2, Bucharest, Romania.
| |
Collapse
|
33
|
Mitkari B, Kerkelä E, Nystedt J, Korhonen M, Jolkkonen J. Unexpected complication in a rat stroke model: exacerbation of secondary pathology in the thalamus by subacute intraarterial administration of human bone marrow-derived mesenchymal stem cells. J Cereb Blood Flow Metab 2015; 35:363-6. [PMID: 25564231 PMCID: PMC4348397 DOI: 10.1038/jcbfm.2014.235] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/28/2014] [Accepted: 11/28/2014] [Indexed: 11/09/2022]
Abstract
This study examined whether human bone marrow mesenchymal stromal/stem cells (BMMSCs) could alleviate the secondary pathology in the thalamus after middle cerebral artery occlusion (MCAO) in rats. Atypical accumulation of both amyloid-β (Aβ) and calcium in the thalamus was significantly higher in rats receiving the BMMSCs infusion 48 hours after MCAO as compared with the vehicle MCAO group. The elevated Aβ/calcium accumulation correlated with the level of impaired sensorimotor function. Although secondary pathology in the thalamus seems to be rodent specific, it needs to be taken into account because it may impair long-term behavioral recovery and negate therapeutic treatment effects.
Collapse
Affiliation(s)
- Bhimashankar Mitkari
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| | - Erja Kerkelä
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Johanna Nystedt
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Matti Korhonen
- Finnish Red Cross Blood Service, Research and Cell Therapy Services, Helsinki, Finland
| | - Jukka Jolkkonen
- Institute of Clinical Medicine-Neurology, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
34
|
Doeppner TR, Hermann DM. Stem cell-based treatments against stroke: observations from human proof-of-concept studies and considerations regarding clinical applicability. Front Cell Neurosci 2014; 8:357. [PMID: 25400548 PMCID: PMC4212679 DOI: 10.3389/fncel.2014.00357] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 10/12/2014] [Indexed: 12/14/2022] Open
Abstract
Ischemic stroke remains a heavy burden for industrialized countries. The only causal therapy is the recanalization of occluded vessels via thrombolysis, which due to a narrow time window still can be offered only to a minority of patients. Since the majority of patients continues to exhibit neurological deficits even following successful thrombolysis, restorative therapies are urgently needed that promote brain remodeling and repair once stroke injury has occurred. Due to their unique properties of action, stem cell-based strategies gained increasing interest during recent years. Using various stroke models in both rodents and primates, the transplantation of stem cells, namely of bone marrow derived mesenchymal stem cells (MSCs) or neural progenitor cells (NPCs), has been shown to promote neurological recovery most likely via indirect bystander actions. In view of promising observations, clinical proof-of-concept studies are currently under way, in which effects of stem and precursor cells are evaluated in human stroke patients. In this review we summarize already published studies, which due to the broad experience in other medical contexts mostly employed bone marrow-derived MSCs by means of intravenous transplantation. With the overall number of clinical trials limited in number, only a fraction of these studies used non-treated control groups, and only single studies were adequately blinded. Despite these limitations, first promising results justify the need for more elaborate clinical trials in order to make stem cell transplantation a success for stroke treatment in the future.
Collapse
Affiliation(s)
- Thorsten R Doeppner
- Department of Neurology, University of Duisburg-Essen Medical School Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University of Duisburg-Essen Medical School Essen, Germany
| |
Collapse
|