1
|
Vaheb S, Afshin S, Ghoshouni H, Ghaffary EM, Farzan M, Shaygannejad V, Thapa S, Zabeti A, Mirmosayyeb O. Neurological efficacy and safety of mesenchymal stem cells (MSCs) therapy in people with multiple sclerosis (pwMS): An updated systematic review and meta-analysis. Mult Scler Relat Disord 2024; 87:105681. [PMID: 38838423 DOI: 10.1016/j.msard.2024.105681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 04/26/2024] [Accepted: 05/10/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Current therapeutic strategies for multiple sclerosis (MS) aim to suppress the immune response and reduce relapse rates. As alternative treatments, mesenchymal stem cells (MSCs) are being explored. MSCs show promise in repairing nerve tissue and reducing autoimmune responses in people with MS (pwMS). OBJECTIVE This review delves into the literature on the efficacy and safety of MSC therapy for pwMS. METHODS A comprehensive search strategy was employed to identify relevant articles from five databases until January 2024. The inclusion criteria encompassed interventional studies. Efficacy and safety data concerning MSC therapy in relapsing-remitting MS (RRMS), secondary progressive MS (SPMS), and primary progressive MS (PPMS) groups were extracted and analyzed. RESULTS A comprehensive analysis encompassing 30 studies revealed that individuals who underwent intrathecal (IT) protocol-based transplantation of MSCs experienced a noteworthy improvement in their expanded disability status scale (EDSS) compared to the placebo group. Weighted mean difference (WMD) was -0.28; 95 % CI -0.53 to -0.03, I2 = 0 %, p-value = 0.028); however, the intravenous (IV) group did not show significant changes in EDSS scores. The annualized relapse rate (ARR) did not significantly decrease among pwMS (WMD = -0.34; 95 % CI -1.05 to 0.38, I2 = 98 %, p-value = 0.357). Favorable results were observed in magnetic resonance imaging (MRI), with only 19.11 % of pwMS showing contrast-enhanced lesions (CEL) in the short term and no long-term MRI activity. The most common complications in both short-term and long-term follow-ups were infection, back pain, and gastrointestinal symptoms. CONCLUSIONS The study highlights the safety potential of MSC therapy for pwMS. While MRI-based neural regeneration shows significant treatment potential, the effectiveness of MSC therapy remains uncertain due to study limitations and ineffective outcome measures. Further research is needed to establish efficacy and optimize evaluation methods for MSC therapy on pwMS.
Collapse
Affiliation(s)
- Saeed Vaheb
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sahra Afshin
- Department of Neurology, School of Medicine, Hormozgan University of Medical Sciences, Bandarabbas, Iran
| | - Hamed Ghoshouni
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Moases Ghaffary
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahour Farzan
- Students Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vahid Shaygannejad
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sangharsha Thapa
- Jacobs School of Biomedical Sciences, University of Buffalo, Department of Neurology, Buffalo, USA
| | - Aram Zabeti
- University of Cincinnati, Cincinnati, OH, USA
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran; Department of Neurology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
2
|
Ghosh S, Bhatti GK, Sharma PK, Kandimalla R, Mastana SS, Bhatti JS. Potential of Nano-Engineered Stem Cells in the Treatment of Multiple Sclerosis: A Comprehensive Review. Cell Mol Neurobiol 2023; 44:6. [PMID: 38104307 DOI: 10.1007/s10571-023-01434-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023]
Abstract
Multiple sclerosis (MS) is a chronic and degrading autoimmune disorder mainly targeting the central nervous system, leading to progressive neurodegeneration, demyelination, and axonal damage. Current treatment options for MS are limited in efficacy, generally linked to adverse side effects, and do not offer a cure. Stem cell therapies have emerged as a promising therapeutic strategy for MS, potentially promoting remyelination, exerting immunomodulatory effects and protecting against neurodegeneration. Therefore, this review article focussed on the potential of nano-engineering in stem cells as a therapeutic approach for MS, focusing on the synergistic effects of combining stem cell biology with nanotechnology to stimulate the proliferation of oligodendrocytes (OLs) from neural stem cells and OL precursor cells, by manipulating neural signalling pathways-PDGF, BMP, Wnt, Notch and their essential genes such as Sox, bHLH, Nkx. Here we discuss the pathophysiology of MS, the use of various types of stem cells in MS treatment and their mechanisms of action. In the context of nanotechnology, we present an overview of its applications in the medical and research field and discuss different methods and materials used to nano-engineer stem cells, including surface modification, biomaterials and scaffolds, and nanoparticle-based delivery systems. We further elaborate on nano-engineered stem cell techniques, such as nano script, nano-exosome hybrid, nano-topography and their potentials in MS. The article also highlights enhanced homing, engraftment, and survival of nano-engineered stem cells, targeted and controlled release of therapeutic agents, and immunomodulatory and tissue repair effects with their challenges and limitations. This visual illustration depicts the process of utilizing nano-engineering in stem cells and exosomes for the purpose of delivering more accurate and improved treatments for Multiple Sclerosis (MS). This approach targets specifically the creation of oligodendrocytes, the breakdown of which is the primary pathological factor in MS.
Collapse
Affiliation(s)
- Sushruta Ghosh
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences Central, University of Punjab, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India
| | - Pushpender Kumar Sharma
- Amity Institute of Biotechnology, Amity University, Rajasthan, India
- Amity Centre for Nanobiotechnology and Nanomedicine, Amity University, Rajasthan, India
| | - Ramesh Kandimalla
- Department of Biochemistry, Kakatiya Medical College, Warangal, Telangana, India
- Department of Applied Biology, CSIR-Indian Institute of Technology, Hyderabad, India
| | - Sarabjit Singh Mastana
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences Central, University of Punjab, Bathinda, India.
| |
Collapse
|
3
|
Huang LH, Pan ZY, Pan YJ, Yang FY. Magnetization transfer ratio for assessing remyelination after transcranial ultrasound stimulation in the lysolecithin rat model of multiple sclerosis. Cereb Cortex 2023; 33:1403-1411. [PMID: 35368059 DOI: 10.1093/cercor/bhac144] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/15/2022] Open
Abstract
It has been shown that transcranial ultrasound stimulation (TUS) is capable of attenuating myelin loss and providing neuroprotection in animal models of brain disorders. In this study, we investigated the ability of TUS to promote remyelination in the lysolecithin (LPC)-induced local demyelination in the hippocampus. Demyelination was induced by the micro-injection of 1.5 μL LPC (1%) into the rat hippocampus and the treated group received daily TUS for 5 or 12 days. Magnetic resonance imaging techniques, including magnetization transfer ratio (MTR) and T2-weighted imaging, were used to longitudinally characterize the demyelination model. Furthermore, the therapeutic effects of TUS on LPC-induced demyelination were assessed by Luxol fast blue (LFB) staining. Our data revealed that reductions in MTR values observed during demyelination recover almost completely upon remyelination. The MTR values in demyelinated lesions were significantly higher in TUS-treated rats than in the LPC-only group after undergoing TUS. Form histological observation, TUS significantly reduced the size of demyelinated lesion 7 days after LPC administration. This study demonstrated that MTR was a sensitive and reproducible quantitative marker to assess remyelination process in vivo during TUS treatment. These findings might open new promising treatment strategies for demyelinating diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Li-Hsin Huang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Taipei 11221, Taiwan
| | - Zih-Yun Pan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Taipei 11221, Taiwan
| | - Yi-Ju Pan
- Department of Psychiatry, Far Eastern Memorial Hospital, No. 21, Sec. 2, Nanya S. Rd., Banciao Dist., New Taipei City 220, Taiwan.,Institute of Public Health, National Yang Ming Chiao Tung University School of Medicine, No. 155, Sec. 2, Li-Nong St., Taipei 11221, Taiwan
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Taipei 11221, Taiwan
| |
Collapse
|
4
|
Genchi A, Brambilla E, Sangalli F, Radaelli M, Bacigaluppi M, Furlan R, Andolfo A, Drago D, Magagnotti C, Scotti GM, Greco R, Vezzulli P, Ottoboni L, Bonopane M, Capilupo D, Ruffini F, Belotti D, Cabiati B, Cesana S, Matera G, Leocani L, Martinelli V, Moiola L, Vago L, Panina-Bordignon P, Falini A, Ciceri F, Uglietti A, Sormani MP, Comi G, Battaglia MA, Rocca MA, Storelli L, Pagani E, Gaipa G, Martino G. Neural stem cell transplantation in patients with progressive multiple sclerosis: an open-label, phase 1 study. Nat Med 2023; 29:75-85. [PMID: 36624312 PMCID: PMC9873560 DOI: 10.1038/s41591-022-02097-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 10/17/2022] [Indexed: 01/11/2023]
Abstract
Innovative pro-regenerative treatment strategies for progressive multiple sclerosis (PMS), combining neuroprotection and immunomodulation, represent an unmet need. Neural precursor cells (NPCs) transplanted in animal models of multiple sclerosis have shown preclinical efficacy by promoting neuroprotection and remyelination by releasing molecules sustaining trophic support and neural plasticity. Here we present the results of STEMS, a prospective, therapeutic exploratory, non-randomized, open-label, single-dose-finding phase 1 clinical trial ( NCT03269071 , EudraCT 2016-002020-86), performed at San Raffaele Hospital in Milan, Italy, evaluating the feasibility, safety and tolerability of intrathecally transplanted human fetal NPCs (hfNPCs) in 12 patients with PMS (with evidence of disease progression, Expanded Disability Status Scale ≥6.5, age 18-55 years, disease duration 2-20 years, without any alternative approved therapy). The safety primary outcome was reached, with no severe adverse reactions related to hfNPCs at 2-year follow-up, clearly demonstrating that hfNPC therapy in PMS is feasible, safe and tolerable. Exploratory secondary analyses showed a lower rate of brain atrophy in patients receiving the highest dosage of hfNPCs and increased cerebrospinal fluid levels of anti-inflammatory and neuroprotective molecules. Although preliminary, these results support the rationale and value of future clinical studies with the highest dose of hfNPCs in a larger cohort of patients.
Collapse
Affiliation(s)
- Angela Genchi
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Elena Brambilla
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sangalli
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Radaelli
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Bacigaluppi
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Roberto Furlan
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Clinical Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Annapaola Andolfo
- grid.18887.3e0000000417581884ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Denise Drago
- grid.18887.3e0000000417581884ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Magagnotti
- grid.18887.3e0000000417581884ProMeFa, Proteomics and Metabolomics Facility, Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Maria Scotti
- grid.18887.3e0000000417581884Center for Omics Sciences (COSR), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raffaella Greco
- grid.18887.3e0000000417581884Haematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Vezzulli
- grid.18887.3e0000000417581884Department of Neuroradiology and CERMAC, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Linda Ottoboni
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Bonopane
- grid.18887.3e0000000417581884Clinical Trial Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Capilupo
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Ruffini
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniela Belotti
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Benedetta Cabiati
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Stefania Cesana
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Giada Matera
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Letizia Leocani
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Vittorio Martinelli
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Moiola
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Vago
- grid.18887.3e0000000417581884Haematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Panina-Bordignon
- grid.18887.3e0000000417581884Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | - Andrea Falini
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Department of Neuroradiology and CERMAC, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Haematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Anna Uglietti
- grid.414818.00000 0004 1757 8749Department of Gynaecology, IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Pia Sormani
- grid.5606.50000 0001 2151 3065Biostatistics Unit, Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy
| | - Giancarlo Comi
- grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy
| | | | - Maria A. Rocca
- grid.18887.3e0000000417581884Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy ,grid.15496.3f0000 0001 0439 0892University Vita-Salute San Raffaele, Milan, Italy ,grid.18887.3e0000000417581884Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Loredana Storelli
- grid.18887.3e0000000417581884Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Pagani
- grid.18887.3e0000000417581884Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giuseppe Gaipa
- grid.415025.70000 0004 1756 8604M. Tettamanti Research Center, Pediatric Clinic University of Milano-Bicocca, San Gerardo Hospital, Monza, Italy ,grid.415025.70000 0004 1756 8604Laboratorio di Terapia Cellulare e Genica Stefano Verri, ASST-Monza, Ospedale San Gerardo, Monza, Italy
| | - Gianvito Martino
- Neuroimmunology Unit, Institute of Experimental Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy. .,Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy. .,University Vita-Salute San Raffaele, Milan, Italy.
| |
Collapse
|
5
|
Ahmed T. Neural stem cell engineering for the treatment of multiple sclerosis. BIOMEDICAL ENGINEERING ADVANCES 2022. [DOI: 10.1016/j.bea.2022.100053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
6
|
Toxicity, Safety, and Efficacy Studies on Mesenchymal Stem Cells Derived from Decidua basalis in Wistar Albino Rats by Intravenous and Subcutaneous Routes. Curr Issues Mol Biol 2022; 44:4045-4058. [PMID: 36135189 PMCID: PMC9498010 DOI: 10.3390/cimb44090277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/13/2022] [Accepted: 08/17/2022] [Indexed: 12/03/2022] Open
Abstract
Ex vivo expanded decidua-basalis(DB)-derived mesenchymal stem cells (MSCs) obtained from single donors have demonstrated therapeutic benefits in in vitro and in vivo studies. In this report, the intravenous and subcutaneous administration of DB-MSCs obtained from five healthy donors was assessed considering clinical grade proliferation, accessibility, and toxic effects in Wistar albino rats. The ability of the obtained DB-MSCs for differentiating, as well as their expression of several cell surface markers and immunomodulatory activities, were all assessed. Clinical standard proliferated cells were administered to animals intravenously and subcutaneously in a series of preclinical models in order to assess their in vivo toxicity, general safety, and tumorigenic possibilities. We established that DB cells exhibit structural and functional traits with MSCs. At various doses supplied intravenously or subcutaneously, the research showed no fatality, abnormal response to therapy, or substantial pathological modifications in the rats. Furthermore, there was no indication of prenatal damage in the same animal species when the rats were repeatedly treated with DBMSCs. Thus, DBMSCs were demonstrated to be non-toxic, non-teratogenic, and non-tumorigenic. To determine whether they can be administrated to human patients without risk, more investigation is recommended.
Collapse
|
7
|
Liu H, Chen B, Zhu Q. Potential application of hydrogel to the diagnosis and treatment of multiple sclerosis. J Biol Eng 2022; 16:10. [PMID: 35395765 PMCID: PMC8991948 DOI: 10.1186/s13036-022-00288-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/12/2022] [Indexed: 11/18/2022] Open
Abstract
Abstract Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system. This disorder may cause progressive and permanent impairment, placing significant physical and psychological strain on sufferers. Each progress in MS therapy marks a significant advancement in neurological research. Hydrogels can serve as a scaffold with high water content, high expansibility, and biocompatibility to improve MS cell proliferation in vitro and therapeutic drug delivery to cells in vivo. Hydrogels may also be utilized as biosensors to detect MS-related proteins. Recent research has employed hydrogels as an adjuvant imaging agent in immunohistochemistry assays. Following an overview of the development and use of hydrogels in MS diagnostic and therapy, this review discussed hydrogel’s advantages and future opportunities in the diagnosis and treatment of MS. Graphical abstract ![]()
Collapse
Affiliation(s)
- Haochuan Liu
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Xiantai Street No. 126, Changchun, TX, 130031, PR China
| | - Bing Chen
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Xiantai Street No. 126, Changchun, TX, 130031, PR China.
| | - Qingsan Zhu
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Xiantai Street No. 126, Changchun, TX, 130031, PR China.
| |
Collapse
|
8
|
Yan Y, Wang X, Zhu G. Endometrium Derived Stem Cells as Potential Candidates in Nervous System Repair. Ann Biomed Eng 2022; 50:485-498. [PMID: 35235077 DOI: 10.1007/s10439-022-02909-0] [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: 08/30/2021] [Accepted: 01/01/2022] [Indexed: 11/24/2022]
Abstract
Limited cell division and lack of endogenous repair mechanisms in the central nervous system, hampers tissue repair following neurodegenerative diseases or tissue injuries. Unlike central nervous system; peripheral nervous system has some capacity to repair after injury, but in case of critical sized defects the use of supporting cells in the neural guidance channels seems inevitable to obtain a satisfactory functional recovery. Stem cell therapies have provided new frontiers in the repair of nervous system largely through paracrine secretion mechanisms. The therapeutic potential of stem cells differs according to their tissue of origin, mode of isolation, administration route, and passage number. During the past decades, studies have been focused on stem cells harvested from disposable tissues such as menstrual blood or biopsies from endometrium. These cells are characterized by their high differentiation and proliferation potential, ease of harvest, and lack of ethical concerns. In the current review, we will discuss the prospects and challenges of endometrial stem cells' application in nervous system repair.
Collapse
Affiliation(s)
- Yifen Yan
- Department of Gynecology, Renmin Hospital, Hubei University of Medicine, Maojian District, No. 39, Chaoyang Zhong Road, Shiyan City, 442000, Hubei Province, China
| | - Xiaoli Wang
- Department of Gynecology, Renmin Hospital, Hubei University of Medicine, Maojian District, No. 39, Chaoyang Zhong Road, Shiyan City, 442000, Hubei Province, China
| | - Guijuan Zhu
- Department of Gynecology, Renmin Hospital, Hubei University of Medicine, Maojian District, No. 39, Chaoyang Zhong Road, Shiyan City, 442000, Hubei Province, China.
| |
Collapse
|
9
|
Clinical application of stem cell therapy in neurogenic bladder: a systematic review and meta-analysis. Int Urogynecol J 2021; 33:2081-2097. [PMID: 34767058 DOI: 10.1007/s00192-021-04986-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 08/23/2021] [Indexed: 01/26/2023]
Abstract
INTRODUCTION AND HYPOTHESIS This review aims to investigate the effect of stem cell (SC) therapy on the management of neurogenic bladder (NGB) in four neurological diseases, including spinal cord injury (SCI), Parkinson's disease (PD), multiple sclerosis (MS), and stroke, in the clinical setting. METHODS An electronic database search was conducted in the Cochrane Library, EMBASE, Proquest, Clinicaltrial.gov , WHO, Google Scholar, MEDLINE via PubMed, Ovid, Web of Science, Scopus, ongoing trial registers, and conference proceedings in June 2019 and updated by hand searching on 1 February 2021. All randomized controlled trials (RCTs), quasi RCTs, phase I/II clinical trials, case-control, retrospective cohorts, and comprehensive case series that evaluated the regenerative potential of SCs on the management of NGB were included. Cochrane appraisal risk of bias checklist and the standardized critical appraisal instrument from the JBI Meta-Analysis of Statistics, Assessment, and Review Instrument (JBI-MAStARI) were used to appraise the studies. RESULTS Twenty-six studies among 1282 relevant publications met our inclusion criteria. Only SC therapy was applied for SCI or MS patients. Phase I/II clinical trials (without control arm) were the most conducted studies, and only four were RCTs. Four studies with 153 participants were included in the meta-analysis. The main route of transplantation was via lumbar puncture. There were no serious adverse events. Only nine studies in SCI and one in MS have used urodynamics, and the others have reported improvement based on patient satisfaction. SC therapy did not significantly improve residual urine volume, detrusor pressure, and maximum bladder capacity. Also, the quality of these publications was low or unclear. CONCLUSION Although most clinical trials provide evidence of the safety and effectiveness of MSCs on the management of NGB, the meta-analysis results did not show a significant improvement; however, the interpretation of study results is difficult because of the lack of placebo controls.
Collapse
|
10
|
Miao J, Ren Z, Zhong Z, Yan L, Xia X, Wang J, Yang J. Mesenchymal Stem Cells: Potential Therapeutic Prospect of Paracrine Pathways in Neonatal Infection. J Interferon Cytokine Res 2021; 41:365-374. [PMID: 34672801 DOI: 10.1089/jir.2021.0094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Infection is the leading cause of admission and mortality in neonatal intensive care units. Immature immune function and antibiotic resistance make the treatment more difficult. However, there is no effective prevention for it. Recently, more and more researches are focusing on stem cell therapy, especially mesenchymal stem cells (MSCs); their potential paracrine effect confer MSCs with a major advantage to treat the immune and inflammatory disorders associated with neonatal infection. In this review, we summarize the basal properties and preclinical evidence of MSCs and explore the potential mechanisms of paracrine factors of MSCs for neonatal infection.
Collapse
Affiliation(s)
- Jiayu Miao
- Department of Pediatrics, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zhuxiao Ren
- Department of Neonatology, and Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Zhicheng Zhong
- Department of Prenatal Diagnosis Center, Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Longli Yan
- Department of Neonatology, and Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xin Xia
- Department of Neonatology, and Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jianlan Wang
- Department of Neonatology, and Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jie Yang
- Department of Neonatology, and Guangdong Women and Children Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
11
|
Hu XM, Zhang Q, Zhou RX, Wu YL, Li ZX, Zhang DY, Yang YC, Yang RH, Hu YJ, Xiong K. Programmed cell death in stem cell-based therapy: Mechanisms and clinical applications. World J Stem Cells 2021; 13:386-415. [PMID: 34136072 PMCID: PMC8176847 DOI: 10.4252/wjsc.v13.i5.386] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 02/06/2023] Open
Abstract
Stem cell-based therapy raises hopes for a better approach to promoting tissue repair and functional recovery. However, transplanted stem cells show a high death percentage, creating challenges to successful transplantation and prognosis. Thus, it is necessary to investigate the mechanisms underlying stem cell death, such as apoptotic cascade activation, excessive autophagy, inflammatory response, reactive oxygen species, excitotoxicity, and ischemia/hypoxia. Targeting the molecular pathways involved may be an efficient strategy to enhance stem cell viability and maximize transplantation success. Notably, a more complex network of cell death receives more attention than one crucial pathway in determining stem cell fate, highlighting the challenges in exploring mechanisms and therapeutic targets. In this review, we focus on programmed cell death in transplanted stem cells. We also discuss some promising strategies and challenges in promoting survival for further study.
Collapse
Affiliation(s)
- Xi-Min Hu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410013, Hunan Province, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Rui-Xin Zhou
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Yan-Lin Wu
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Zhi-Xin Li
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Dan-Yi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Yi-Chao Yang
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| | - Rong-Hua Yang
- Department of Burns, Fo Shan Hospital of Sun Yat-Sen University, Foshan 528000, Guangdong Province, China
| | - Yong-Jun Hu
- Department of Cardiovascular Medicine, Hunan People's Hospital (the First Affiliated Hospital of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Sciences, Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
12
|
Jarrin S, Cabré S, Dowd E. The potential of biomaterials for central nervous system cellular repair. Neurochem Int 2021; 144:104971. [PMID: 33515647 DOI: 10.1016/j.neuint.2021.104971] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 01/01/2023]
Abstract
The central nervous system (CNS) can be injured or damaged through a variety of insults including traumatic injury, stroke, and neurodegenerative or demyelinating diseases, including Alzheimer's disease, Parkinson's disease and multiple sclerosis. Existing pharmacological and other therapeutics strategies are limited in their ability to repair or regenerate damaged CNS tissue meaning there are significant unmet clinical needs facing patients suffering CNS damage and/or degeneration. Through a variety of mechanisms including neuronal replacement, secretion of therapeutic factors, and stimulation of host brain plasticity, cell-based repair offers a potential mechanism to repair and heal the damaged CNS. However, over the decades of its evolution as a therapeutic strategy, cell-based CNS repair has faced significant hurdles that have prevented its translation to widespread clinical practice. In recent years, advances in cell technologies combined with advances in biomaterial-based regenerative medicine and tissue engineering have meant there is very real potential for many of these hurdles to be overcome. This review will provide an overview of the main CNS conditions that lend themselves to cellular repair and will then outline the potential of biomaterial-based approaches for improving the outcome of cellular repair in these conditions.
Collapse
Affiliation(s)
- Sarah Jarrin
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland
| | - Sílvia Cabré
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland; APC Microbiome Ireland, University College Cork, Ireland
| | - Eilís Dowd
- Pharmacology & Therapeutics and Galway Neuroscience Centre, National University of Ireland, Galway, Ireland.
| |
Collapse
|
13
|
Barati S, Tahmasebi F, Faghihi F. Effects of mesenchymal stem cells transplantation on multiple sclerosis patients. Neuropeptides 2020; 84:102095. [PMID: 33059244 DOI: 10.1016/j.npep.2020.102095] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 09/12/2020] [Accepted: 09/13/2020] [Indexed: 02/06/2023]
Abstract
Multiple Sclerosis (MS) is a demyelinating autoimmune disease of the central nervous system (CNS) with symptoms such as neuroinflammation and axonal degeneration. Existing drugs help reduce inflammatory conditions and protect CNS from demyelination and axonal damage; however, these drugs are unable to enhance axonal repair and remyelination. In this regard, cell therapy is considered as a promising regenerative approach to MS treatment. High immunomodulatory capacity, neuro-differentiation and neuroprotection properties have made Mesenchymal Stem Cells (MSCs) particularly useful for regenerative medicine. There are scant studies on the role of MSCs in patients suffering from MS. The low number of MS patients and the lack of control groups in these studies may explain the lack of beneficial effects of MSC transplantation in cell therapies. In this review, we evaluated the beneficial effects of MSC transplantation in clinical studies in terms of immunomodulatory, remyelinating and neuroprotecting properties of MSCs.
Collapse
Affiliation(s)
- Shirin Barati
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Tahmasebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faeze Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Padnahad Co.Ltd, Tehran, Iran.
| |
Collapse
|
14
|
Metzger-Peter K, Kremer LD, Edan G, Loureiro De Sousa P, Lamy J, Bagnard D, Mensah-Nyagan AG, Tricard T, Mathey G, Debouverie M, Berger E, Kerbrat A, Meyer N, De Seze J, Collongues N. The TOTEM RRMS (Testosterone Treatment on neuroprotection and Myelin Repair in Relapsing Remitting Multiple Sclerosis) trial: study protocol for a randomized, double-blind, placebo-controlled trial. Trials 2020; 21:591. [PMID: 32600454 PMCID: PMC7322908 DOI: 10.1186/s13063-020-04517-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 06/15/2020] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Central nervous system damage in multiple sclerosis (MS) is responsible for serious deficiencies. Current therapies are focused on the treatment of inflammation; however, there is an urgent need for innovative therapies promoting neuroregeneration, particularly myelin repair. It is demonstrated that testosterone can act through neural androgen receptors and several clinical observations stimulated an interest in the potential protective effects of testosterone treatment for MS. Here, we sought to demonstrate the effects of a testosterone supplementation in testosterone-deficient men with relapsing-remitting MS. METHODS/DESIGN This report presents the rationale and methodology of TOTEM RRMS, a French, phase 2, multicenter, randomized, placebo-controlled, and double-blind trial, which aims to prevent the progression of MS in men with low testosterone levels by administration of testosterone undecanoate, who were kept under natalizumab (Tysabri®) to overcome the anti-inflammatory effect of testosterone. Forty patients will be randomized into two groups receiving either a testosterone treatment (Nebido®) or a matching placebo. The intervention period for each group will last 66 weeks (treatment will be injected at baseline, week 6, and then every 12 weeks). The main objective is to determine the neuroprotective and remyelinating effects of testosterone using tensor diffusion imaging techniques and thalamic atrophy analyses. As secondary objectives, impacts of the testosterone supplementation will be studied using other conventional and unconventional MRI parameters and with clinical outcomes. DISCUSSION The action of testosterone is observed in different experimental autoimmune encephalomyelitis models and epidemiological studies in humans. However, despite several preclinical data and some small clinical trials in MS, clear evidence for a therapeutic effect of hormone therapy is still missing. Therefore, our goal is to demonstrate the effects of testosterone therapies in MS. As there is no effective treatment currently available on fatigue in MS, careful attention should also be paid to secondary endpoints: fatigue, cognitive functions, and other symptoms that may improve life quality. Assuming a positive outcome of the trial, this treatment could be considered as a new neuroprotective and remyelinating therapy in relapsing-remitting MS and could be applicable to other demyelinating diseases. TRIAL REGISTRATION ClinicalTrials.gov NCT03910738. Registered on 10 April 2019.
Collapse
Affiliation(s)
| | - Laurent Daniel Kremer
- Departement of Neurology, Hôpital de Hautepierre, University Hospital of Strasbourg, Strasbourg, France
| | - Gilles Edan
- Departement of Neurology, Hôpital Pontchaillou, University Hospital of Rennes, Rennes, France
| | - Paulo Loureiro De Sousa
- Laboratory of Engineering Sciences, Computer Science and Imagery (ICube), CNRS, Institute of Biological Physics, University of Strasbourg, Strasbourg, France
| | - Julien Lamy
- Laboratory of Engineering Sciences, Computer Science and Imagery (ICube), CNRS, Institute of Biological Physics, University of Strasbourg, Strasbourg, France
| | - Dominique Bagnard
- Departement of Myelin Biopathology, Neuroprotection and Therapeutic Strategies, UMR_S Inserm 1119, Strasbourg, France
| | - Ayikoe-Guy Mensah-Nyagan
- Departement of Myelin Biopathology, Neuroprotection and Therapeutic Strategies, UMR_S Inserm 1119, Strasbourg, France
| | - Thibault Tricard
- Departement of Urological Surgery, Nouvel Hôpital Civil, University Hospital of Strasbourg, Strasbourg, France
| | - Guillaume Mathey
- Departement of Neurology, Hôpital Central, University Hospital of Nancy, Nancy, France
| | - Marc Debouverie
- Departement of Neurology, Hôpital Central, University Hospital of Nancy, Nancy, France
| | - Eric Berger
- Departement of Neurology, Hôpital Jean Minjoz, University Hospital of Besançon, Besançon, France
| | - Anne Kerbrat
- Department of Neurology, Hôpital de Pontchaillou, University Hospital of Rennes, Rennes, France
| | - Nicolas Meyer
- Departement of Public Health, GMRC University Hospital of Strasbourg, Strasbourg, France
| | - Jérôme De Seze
- Centre d᾿Investigation Clinique INSERM 1434, Strasbourg, France.,Departement of Neurology, Hôpital de Hautepierre, University Hospital of Strasbourg, Strasbourg, France
| | - Nicolas Collongues
- Centre d᾿Investigation Clinique INSERM 1434, Strasbourg, France.,Departement of Neurology, Hôpital de Hautepierre, University Hospital of Strasbourg, Strasbourg, France
| |
Collapse
|
15
|
Mehdipour A, Ebrahimi A, Shiri-Shahsavar MR, Soleimani-Rad J, Roshangar L, Samiei M, Ebrahimi-Kalan A. The potentials of umbilical cord-derived mesenchymal stem cells in the treatment of multiple sclerosis. Rev Neurosci 2020; 30:857-868. [PMID: 31026226 DOI: 10.1515/revneuro-2018-0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/15/2019] [Indexed: 12/12/2022]
Abstract
Stem cell therapy has indicated a promising treatment capacity for tissue regeneration. Multiple sclerosis is an autoimmune-based chronic disease, in which the myelin sheath of the central nervous system is destructed. Scientists have not discovered any cure for multiple sclerosis, and most of the treatments are rather palliative. The pursuit of a versatile treatment option, therefore, seems essential. The immunoregulatory and non-chronic rejection characteristics of mesenchymal stem cells, as well as their homing properties, recommend them as a prospective treatment option for multiple sclerosis. Different sources of mesenchymal stem cells have distinct characteristics and functional properties; in this regard, choosing the most suitable cell therapy approach seems to be challenging. In this review, we will discuss umbilical cord/blood-derived mesenchymal stem cells, their identified exclusive properties compared to another adult mesenchymal stem cells, and the expectations of their potential roles in the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Ahmad Mehdipour
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayyub Ebrahimi
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Haliç University, Istanbul, Turkey
| | | | - Jafar Soleimani-Rad
- Department of Anatomical Sciences, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Department of Anatomical Sciences, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Samiei
- Endodontics Department of Dental Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Radiology, School of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran,
| |
Collapse
|
16
|
Borhani-Haghighi M, Mohamadi Y. Intranasal administration of conditioned medium derived from mesenchymal stem cells-differentiated oligodendrocytes ameliorates experimental autoimmune encephalomyelitis. J Chem Neuroanat 2020; 106:101792. [PMID: 32353514 DOI: 10.1016/j.jchemneu.2020.101792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/05/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022]
Abstract
In multiple sclerosis, myelin sheaths around the axons are degenerated due to uncontrolled inflammation in the central nervous system. Oligodendrocytes (OLs) are myelin-forming cells that secrete trophic factors necessary for myelin protection. Beneficial features of conditioned medium (CM) derived from different stem cells are nowadays under investigation in treating neurodegenerative diseases. Here, we used the differentiation capacity of Wharton's jelly mesenchymal stem cells (WJMSCs) to obtain OLs. Then, the study aimed to evaluate the status of inflammation and myelination in male experimental autoimmune encephalomyelitis (EAE) mice after intranasal administration of CM derived from OLs (OL-CM). Inflammation was studied by evaluating gliosis, inflammatory cell infiltration and expression of inflammation indicators including NLRP3 inflammasome, interleukin-1β, interleukin-18, glial fibrillary acidic protein, and ionized calcium binding adaptor molecule 1. Remyelination was studied by luxol fast blue staining and evaluating the expression of myelin indicators including myelin basic protein and oligodendrocyte transcription factor. In addition, we followed the trend of body weight and functional recovery during the 28-day study. ELISA assay revealed that OL-CM contained brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, and ciliary neurotrophic factor. Data showed that OL-CM moderated inflammation, augmented remyelination, and gained normal body weight. Notably, these anti-inflammatory and regenerative effects of OL-CM improved neurological functions in EAE mice. In conclusion, the current study offered a new choice for treating multiple sclerosis using noninvasive intranasal administration of CM harvested from easily achievable WJMSCs-differentiated OLs.
Collapse
Affiliation(s)
- Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Mohamadi
- Department of Anatomy, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran.
| |
Collapse
|
17
|
Ganji R, Razavi S, Ghasemi N, Mardani M. Improvement of Remyelination in Demyelinated Corpus Callosum Using Human Adipose-Derived Stem Cells (hADSCs) and Pregnenolone in the Cuprizone Rat Model of Multiple Sclerosis. J Mol Neurosci 2020; 70:1088-1099. [PMID: 32314194 DOI: 10.1007/s12031-020-01515-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 02/19/2020] [Indexed: 01/24/2023]
Abstract
Adipose-derived stem cells (ASCs) have neuroprotective effects, and their repair ability has been approved in neurodegenerative studies. Pregnenolone as a neurosteroid plays significant roles in neurogenesis. We aimed to consider the effect of ADSCs and pregnenolone injection on the multiple sclerosis (MS) model created by cuprizone. Male Wistar rats (n = 36) were fed with an ordinary diet or a diet with cuprizone (0.6%) for 3 weeks. H-ADSCs were taken from patients with lipoaspirate surgery. The rats were divided into six groups (n = 6): healthy, MS, sham, pregnenolone injection, ADSCs injection, pregnenolone and ADSCs injection. Behavioral test, histological examination and TEM were conducted. The specific markers for myelin and cell differentiation were assessed using immunohistochemistry staining. Additionally, the measure of MBP and MOG gene expression and the amount of related proteins were determined using real-time RT-PCR and ELISA techniques, respectively. Histologic results showed that induced demyelination in corpus callosum fibers. TEM revealed an increased thickness of myelin in fibers in the treated groups (P < 0.05). Injection of hADSC and pregnenolone significantly increased the expression levels of MBP and MOG (P < 0.001). The mean percentage of MOG and MBP markers were significantly increased in the treated groups compared to MS and sham groups (P < 0.05). Moreover, the OD level of MBP and MOG proteins showed that their values in the ADSCs/pregnenolone group were close to those of the control group without a significant difference. Our data indicated the remyelination potency and cell differentiation can improve with ADSCs and pregnenolone treatments in the multiple sclerosis model which created by cuprizone in rats.
Collapse
Affiliation(s)
- Rasoul Ganji
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 81744-176, Iran
| | - Shahnaz Razavi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 81744-176, Iran.
| | - Nazem Ghasemi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 81744-176, Iran
| | - Mohammad Mardani
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 81744-176, Iran.
| |
Collapse
|
18
|
Thomas AM, Li S, Chu C, Shats I, Xu J, Calabresi PA, van Zijl PCM, Walczak P, Bulte JWM. Evaluation of cell transplant-mediated attenuation of diffuse injury in experimental autoimmune encephalomyelitis using onVDMP CEST MRI. Exp Neurol 2020; 329:113316. [PMID: 32304749 DOI: 10.1016/j.expneurol.2020.113316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022]
Abstract
The development and translation of cell therapies have been hindered by an inability to predict and evaluate their efficacy after transplantation. Using an experimental autoimmune encephalomyelitis (EAE) mouse model of multiple sclerosis (MS), we studied attenuation of the diffuse injury characteristic of EAE and MS by transplanted glial-restricted precursor cells (GRPs). We assessed the potential of on-resonance variable delay multiple pulse (onVDMP) chemical exchange saturation transfer (CEST) MRI to visualize this attenuation. Allogeneic GRPs transplanted in the motor cortex or lateral ventricles attenuated paralysis in EAE mice and attenuated differences compared to naïve mice in onVDMP CEST signal 5 days after transplantation near the transplantation site. Histological analysis revealed that transplanted GRPs co-localized with attenuated astrogliosis. Hence, diffuse injury-sensitive onVDMP CEST MRI may complement conventional MRI to locate and monitor tissue regions responsive to GRP therapy.
Collapse
Affiliation(s)
- A M Thomas
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, United States of America; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, United States of America
| | - S Li
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, United States of America; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, United States of America
| | - C Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, United States of America; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, United States of America
| | - I Shats
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, United States of America; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, United States of America
| | - J Xu
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, United States of America
| | - P A Calabresi
- Department of Neurology, The Johns Hopkins University School of Medicine, United States of America; The Solomon H Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, United States of America
| | - P C M van Zijl
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, United States of America; Department of Oncology, the Johns Hopkins University School of Medicine, United States of America
| | - P Walczak
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, United States of America; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, United States of America
| | - J W M Bulte
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, United States of America; Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, United States of America; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, United States of America; Department of Oncology, the Johns Hopkins University School of Medicine, United States of America; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, United States of America; Department of Chemical & Biomolecular Engineering, The Johns Hopkins University School of Medicine, United States of America.
| |
Collapse
|
19
|
Chu DT, Phuong TNT, Tien NLB, Tran DK, Thanh VV, Quang TL, Truong DT, Pham VH, Ngoc VTN, Chu-Dinh T, Kushekhar K. An Update on the Progress of Isolation, Culture, Storage, and Clinical Application of Human Bone Marrow Mesenchymal Stem/Stromal Cells. Int J Mol Sci 2020; 21:E708. [PMID: 31973182 PMCID: PMC7037097 DOI: 10.3390/ijms21030708] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/13/2022] Open
Abstract
Bone marrow mesenchymal stem/stromal cells (BMSCs), which are known as multipotent cells, are widely used in the treatment of various diseases via their self-renewable, differentiation, and immunomodulatory properties. In-vitro and in-vivo studies have supported the understanding mechanisms, safety, and efficacy of BMSCs therapy in clinical applications. The number of clinical trials in phase I/II is accelerating; however, they are limited in the size of subjects, regulations, and standards for the preparation and transportation and administration of BMSCs, leading to inconsistency in the input and outcome of the therapy. Based on the International Society for Cellular Therapy guidelines, the characterization, isolation, cultivation, differentiation, and applications can be optimized and standardized, which are compliant with good manufacturing practice requirements to produce clinical-grade preparation of BMSCs. This review highlights and updates on the progress of production, as well as provides further challenges in the studies of BMSCs, for the approval of BMSCs widely in clinical application.
Collapse
Affiliation(s)
- Dinh-Toi Chu
- Faculty of Biology, Hanoi National University of Education, Hanoi 100000, Vietnam
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam;
| | - Thuy Nguyen Thi Phuong
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju 61186, Korea
| | - Nguyen Le Bao Tien
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam; (N.L.B.T.); (V.V.T.)
| | - Dang Khoa Tran
- Department of Anatomy, University of Medicine Pham Ngoc Thach, Ho Chi Minh City 700000, Vietnam;
| | - Vo Van Thanh
- Institute of Orthopaedics and Trauma Surgery, Viet Duc Hospital, Hanoi 100000, Vietnam; (N.L.B.T.); (V.V.T.)
- Department of Surgery, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Thuy Luu Quang
- Center for Anesthesia and Surgical Intensive Care, Viet Duc Hospital, Hanoi 100000, Vietnam;
| | | | - Van Huy Pham
- AI Lab, Faculty of Information Technology, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam
| | - Vo Truong Nhu Ngoc
- School of Odonto Stomatology, Hanoi Medical University, Hanoi 100000, Vietnam;
| | - Thien Chu-Dinh
- Institute for Research and Development, Duy Tan University, Danang 550000, Vietnam
| | - Kushi Kushekhar
- Institute of Cancer Research, Oslo University Hospital, 0310 Oslo, Norway;
| |
Collapse
|
20
|
Abstract
Abstract
The umbilical cord is an unlimited source of mesenchymal stem cells (MSC) and hematopoietic stem cells (HSC). MSC obtained from the umbilical cord can be differentiated into different types of mesodermal cells, e.g. chondrocytes, osteocytes, adipocytes, and myocytes. It is also worth mentioning that there are reports of MSC differentiation into endo and ectodermal cells. The immunosuppressive properties of MSCs can protect against graft versus host disease as well as prevent rejection after transplantation. Umbilical cord stem cells can be frozen and then stored in liquid nitrogen for many years. In this work, we focused on the use of preclinical and clinical umbilical cord stem cells in disease entities such as type I diabetes, chronic renal failure, and multiple sclerosis. Furthermore, the anti-cancer properties of Wharton’s jelly cells are described.
Running title: Umbilical cord stem cells in humans
Collapse
|
21
|
Mansoor SR, Zabihi E, Ghasemi-Kasman M. The potential use of mesenchymal stem cells for the treatment of multiple sclerosis. Life Sci 2019; 235:116830. [PMID: 31487529 DOI: 10.1016/j.lfs.2019.116830] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 08/23/2019] [Accepted: 09/01/2019] [Indexed: 12/21/2022]
Abstract
Multiple sclerosis (MS) is a chronic autoimmune inflammatory disease of the central nervous system (CNS). In attempt to identify an appropriate treatment for improving the neurological symptoms and remyelination process, autologous and allogenic transplantation of mesenchymal stem cells (MSCs) have been introduced as an effective therapeutic strategy in MS. MSCs are a heterogeneous subset of pluripotent non-hematopoietic stromal cells that are isolated from bone marrow, adipose tissue, placenta and other sources. MSCs have considerable therapeutic effects due to their ability in differentiation, migration, immune-modulation and neuroregeneration. To date, numerous experimental and clinical studies demonstrated that MSCs therapy improves the CNS repair and modulates functional neurological symptoms. Here, we provided an overview of the current knowledge about the clinical applications of MSCs in MS. Furthermore, the major challenges and risks of MSCs therapy in MS patients have been elucidated.
Collapse
Affiliation(s)
- Sahar Rostami Mansoor
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ebrahim Zabihi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Neuroscience Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
22
|
Ge S, Jiang X, Paul D, Song L, Wang X, Pachter JS. Human ES-derived MSCs correct TNF-α-mediated alterations in a blood-brain barrier model. Fluids Barriers CNS 2019; 16:18. [PMID: 31256757 PMCID: PMC6600885 DOI: 10.1186/s12987-019-0138-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
Background Immune cell trafficking into the CNS is considered to contribute to pathogenesis in MS and its animal model, EAE. Disruption of the blood–brain barrier (BBB) is a hallmark of these pathologies and a potential target of therapeutics. Human embryonic stem cell-derived mesenchymal stem/stromal cells (hES-MSCs) have shown superior therapeutic efficacy, compared to bone marrow-derived MSCs, in reducing clinical symptoms and neuropathology of EAE. However, it has not yet been reported whether hES-MSCs inhibit and/or repair the BBB damage associated with neuroinflammation that accompanies EAE. Methods BMECs were cultured on Transwell inserts as a BBB model for all the experiments. Disruption of BBB models was induced by TNF-α, a pro-inflammatory cytokine that is a hallmark of acute and chronic neuroinflammation. Results Results indicated that hES-MSCs reversed the TNF-α-induced changes in tight junction proteins, permeability, transendothelial electrical resistance, and expression of adhesion molecules, especially when these cells were placed in direct contact with BMEC. Conclusions hES-MSCs and/or products derived from them could potentially serve as novel therapeutics to repair BBB disturbances in MS. Electronic supplementary material The online version of this article (10.1186/s12987-019-0138-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shujun Ge
- Blood-Brain Barrier Laboratory, Dept. of Immunology, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA.
| | - Xi Jiang
- Blood-Brain Barrier Laboratory, Dept. of Immunology, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA.,Perelman School of Medicine at University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Debayon Paul
- Blood-Brain Barrier Laboratory, Dept. of Immunology, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Li Song
- ImStem Biotechnology, Inc., 400 Farmington Ave., Farmington, CT, 06030, USA
| | - Xiaofang Wang
- ImStem Biotechnology, Inc., 400 Farmington Ave., Farmington, CT, 06030, USA
| | - Joel S Pachter
- Blood-Brain Barrier Laboratory, Dept. of Immunology, UConn Health, 263 Farmington Ave, Farmington, CT, 06030, USA
| |
Collapse
|
23
|
't Hart BA. Experimental autoimmune encephalomyelitis in the common marmoset: a translationally relevant model for the cause and course of multiple sclerosis. Primate Biol 2019; 6:17-58. [PMID: 32110715 PMCID: PMC7041540 DOI: 10.5194/pb-6-17-2019] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
Aging Western societies are facing an increasing prevalence of chronic
autoimmune-mediated inflammatory disorders (AIMIDs) for which treatments that are safe and effective are scarce. One of the
main reasons for this situation is the lack of animal models, which accurately replicate
clinical and pathological aspects of the human diseases. One important AIMID is the
neuroinflammatory disease multiple sclerosis (MS), for which the mouse experimental
autoimmune encephalomyelitis (EAE) model has been frequently used in preclinical
research. Despite some successes, there is a long list of experimental treatments that
have failed to reproduce promising effects observed in murine EAE models when they were
tested in the clinic. This frustrating situation indicates a wide validity gap between
mouse EAE and MS. This monography describes the development of an EAE model in nonhuman
primates, which may help to bridge the gap.
Collapse
Affiliation(s)
- Bert A 't Hart
- Department of Immunobiology, Biomedical Primate Research Centre, Rijswijk, the Netherlands.,Department of Biomedical Sciences of Cells and Systems, University Medical Center Groningen, the Netherlands
| |
Collapse
|
24
|
Genetically modified hematopoietic stem/progenitor cells that produce IL-10-secreting regulatory T cells. Proc Natl Acad Sci U S A 2019; 116:2634-2639. [PMID: 30683721 DOI: 10.1073/pnas.1811984116] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Random amino acid copolymers used in the treatment of multiple sclerosis in man or experimental autoimmune encephalomyelitis (EAE) in mice [poly(Y,E,A,K)n, known as Copaxone, and poly(Y,F,A,K)n] function at least in part by generation of IL-10-secreting regulatory T cells that mediate bystander immunosuppression. The mechanism through which these copolymers induce Tregs is unknown. To investigate this question, four previously described Vα3.2 Vβ14 T cell receptor (TCR) cDNAs, the dominant clonotype generated in splenocytes after immunization of SJL mice, that differed only in their CDR3 sequences were utilized to generate retrogenic mice. The high-level production of IL-10 as well as IL-5 and small amounts of the related cytokines IL-4 and IL-13 by CD4+ T cells isolated from the splenocytes of these mice strongly suggests that the TCR itself encodes information for specific cytokine secretion. The proliferation and production of IL-10 by these Tregs was costimulated by activation of glucocorticoid-induced TNF receptor (GITR) (expressed at high levels by these cells) through its ligand GITRL. A mechanism for generation of cells with this specificity is proposed. Moreover, retrogenic mice expressing these Tregs were protected from induction of EAE by the appropriate autoantigen.
Collapse
|
25
|
Dupont G, Yilmaz E, Loukas M, Macchi V, De Caro R, Tubbs RS. Human embryonic stem cells: Distinct molecular personalities and applications in regenerative medicine. Clin Anat 2018; 32:354-360. [PMID: 30521112 PMCID: PMC6850663 DOI: 10.1002/ca.23318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 12/02/2018] [Indexed: 12/11/2022]
Abstract
The field of stem cell biology is exciting because it provides researchers and clinicians with seemingly unlimited applications for treating many human diseases. Stem cells are a renewable source of pluripotent cells that can differentiate into nearly all human cell types. In this article we focus particularly on human embryonic stem (hES) cells, derived from the inner cell mass of the blastocyst and cultured for expansion while remaining undifferentiated, to explore their unique molecular personalities and clinical applications. The aim of this literature review is to reflect the interest in hES cells and to provide a resource for researchers and clinicians interested in the molecular characteristics of such cells. Clin. Anat. 32:354–360, 2019. © 2018 The Authors. Clinical Anatomy published by Wiley Periodicals, Inc. on behalf of American Association of Clinical Anatomists.
Collapse
Affiliation(s)
| | - Emre Yilmaz
- Seattle Science Foundation, Seattle, Washington
| | - Marios Loukas
- Department of Anatomical Sciences, St. George's University, Grenada, West Indies
| | - Veronica Macchi
- Department of Neuroscience, Anatomy Institute, University of Padova, Padova, Italy
| | - Raffaele De Caro
- Department of Neuroscience, Anatomy Institute, University of Padova, Padova, Italy
| | - R Shane Tubbs
- Seattle Science Foundation, Seattle, Washington.,Department of Anatomical Sciences, St. George's University, Grenada, West Indies
| |
Collapse
|
26
|
Fazeli Z, Abedindo A, Omrani MD, Ghaderian SMH. Mesenchymal Stem Cells (MSCs) Therapy for Recovery of Fertility: a Systematic Review. Stem Cell Rev Rep 2018; 14:1-12. [PMID: 28884412 DOI: 10.1007/s12015-017-9765-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In recent years, the mesenchymal stem cells (MSCs) have provided the new opportunities to treat different disorders including infertility. Different studies have suggested that the MSCs have ability to differentiate into germ-like cells under specific induction conditions as well as transplantation to gonadal tissues. The aim of this systematic review was to evaluate the results obtained from different studies on MSCs therapy for promoting fertility. This search was done in PubMed and Science Direct databases using key words MSCs, infertility, therapy, germ cell, azoospermia, ovarian failure and mesenchymal stem cell. Among the more than 11,400 papers, 53 studies were considered eligible for more evaluations. The obtained results indicated that the most studies were performed on MSCs derived from bone marrow and umbilical cord as compared with the other types of MSCs. Different evaluations on animal models as well as in vitro studies supported from their role in the recovery of spermatogenesis and folliculogenesis. Although the data obtained from this systematic review are promising, but the further studies need to assess the efficiency and safety of transplantation of these cells in fertility recovery.
Collapse
Affiliation(s)
- Zahra Fazeli
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Atieh Abedindo
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, No 23, Shahid Labbafi Nejad Educational Hospital, Amir Ebrahimi St, Pasdaran Ave, Tehran, Iran
| | - Sayyed Mohammad Hossein Ghaderian
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, No 23, Shahid Labbafi Nejad Educational Hospital, Amir Ebrahimi St, Pasdaran Ave, Tehran, Iran
| |
Collapse
|
27
|
He JG, Xie QL, Li BB, Zhou L, Yan D. Exosomes Derived from IDO1-Overexpressing Rat Bone Marrow Mesenchymal Stem Cells Promote Immunotolerance of Cardiac Allografts. Cell Transplant 2018; 27:1657-1683. [PMID: 30311501 PMCID: PMC6299201 DOI: 10.1177/0963689718805375] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background: The immunosuppressive activity of mesenchymal stem cells (MSCs) has been exploited to induce tolerance after organ transplantation. The indoleamine 2,3-dioxygenase (IDO) may have beneficial effects in the immunoregulatory properties of MSCs. It was recently revealed that exosomes derived from MSCs play important roles in mediating the biological functions of MSCs. This study aimed to explore the roles of exosomes derived from MSCs in the induction of immune tolerance. Methods: Dendritic cells (DCs) and T-cells were cultured with exosomes derived from rat bone marrow MSCs (BMSCs) overexpressing IDO1 or controls. For the in-vivo study, rats received heart transplants and were treated with exosomes from IDO-BMSCs and heart function was evaluated. Flow cytometry was used to detect expression of cell surface markers. Cytokine levels were detected in culture supernatants or serum samples. Protein and microRNA expressions in exosomes were investigated by chips. Results: Exosomes from IDO-BMSCs cultured with DCs and T-cells (1) downregulated CD40, CD86, CD80, MHC-II, CD45RA, CD45RA+CD45RB, OX62, and upregulated CD274 expression, (2) increased the number of regulatory T-cells (Tregs) and decreased the number of CD8+ T-cells, and (3) decreased the levels of pro-inflammatory cytokines, but increased the levels of anti-inflammatory cytokines compared with the other groups. Transplanted rats, which were injected with exosomes from IDO-BMSCs, had reduced allograft-targeting immune responses and improved cardiac allograft function. Exosomes secreted by IDO-BMSCs exhibited significant upregulations of the immunoregulatory protein FHL-1, miR-540-3p, and a downregulation of miR-338-5p. Conclusion: Exosomes derived from IDO-BMSCs can be used to promote immunotolerance and prolong the survival of cardiac allografts.
Collapse
Affiliation(s)
- Ji-Gang He
- Department of Cardiovascular Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Qiao-Li Xie
- Department of Cardiovascular Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Bei-Bei Li
- Department of Cardiovascular Surgery, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Liang Zhou
- Department of Cardiology, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, China
| | - Dan Yan
- Department of Intensive Care Unit, First People's Hospital of Yunnan Province, Yunnan Province, China
| |
Collapse
|
28
|
Wang S, Zhu R, Li H, Li J, Han Q, Zhao RC. Mesenchymal stem cells and immune disorders: from basic science to clinical transition. Front Med 2018; 13:138-151. [PMID: 30062557 DOI: 10.1007/s11684-018-0627-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/12/2017] [Indexed: 02/08/2023]
Abstract
As a promising candidate seed cell type in regenerative medicine, mesenchymal stem cells (MSCs) have attracted considerable attention. The unique capacity of MSCs to exert a regulatory effect on immunity in an autologous/allergenic manner makes them an attractive therapeutic cell type for immune disorders. In this review, we discussed the current knowledge of and advances in MSCs, including its basic biological properties, i.e., multilineage differentiation, secretome, and immunomodulation. Specifically, on the basis of our previous work, we proposed three new concepts of MSCs, i.e., "subtotipotent stem cell" hypothesis, MSC system, and "Yin and Yang" balance of MSC regulation, which may bring new insights into our understanding of MSCs. Furthermore, we analyzed data from the Clinical Trials database ( http://clinicaltrials.gov ) on registered clinical trials using MSCs to treat a variety of immune diseases, such as graft-versus-host disease, systemic lupus erythematosus, and multiple sclerosis. In addition, we highlighted MSC clinical trials in China and discussed the challenges and future directions in the field of MSC clinical application.
Collapse
Affiliation(s)
- Shihua Wang
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Rongjia Zhu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Hongling Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Jing Li
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Qin Han
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China.,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.,Peking Union Medical College Hospital, Beijing, 100005, China.,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, 100005, China. .,School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China. .,Peking Union Medical College Hospital, Beijing, 100005, China. .,Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences, Beijing, 100005, China.
| |
Collapse
|
29
|
Xu L, Liu Y, Sun Y, Wang B, Xiong Y, Lin W, Wei Q, Wang H, He W, Wang B, Li G. Tissue source determines the differentiation potentials of mesenchymal stem cells: a comparative study of human mesenchymal stem cells from bone marrow and adipose tissue. Stem Cell Res Ther 2017; 8:275. [PMID: 29208029 PMCID: PMC5718061 DOI: 10.1186/s13287-017-0716-x] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 08/09/2017] [Accepted: 10/30/2017] [Indexed: 12/20/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) possess intrinsic regeneration capacity as part of the repair process in response to injury, such as fracture or other tissue injury. Bone marrow and adipose tissue are the major sources of MSCs. However, which cell type is more effective and suitable for cell therapy remains to be answered. The intrinsic molecular mechanism supporting the assertion has also been lacking. Methods Human bone marrow-derived MSCs (BMSCs) and adipose tissue-derived MSCs (ATSCs) were isolated from bone marrow and adipose tissue obtained after total hip arthroplasty. ATSCs and BMSCs were incubated in standard growth medium. Trilineage differentiation including osteogenesis, adipogenesis, and chondrogenesis was performed by addition of relevant induction mediums. The expression levels of trilineage differentiation marker genes were evaluated by quantitative RT-PCR. The methylation status of CpG sites of Runx2, PPARγ, and Sox9 promoters were checked by bisulfite sequencing. In addition, ectopic bone formation and calvarial bone critical defect models were used to evaluate the bone regeneration ability of ATSCs and BMSCs in vivo. Results The results showed that BMSCs possessed stronger osteogenic and lower adipogenic differentiation potentials compared to ATSCs. There was no significant difference in the chondrogenic differentiation potential. The CpG sites of Runx2 promoter in BMSCs were hypomethylated, while in ATSCs they were hypermethylated. The CpG sites of PPARγ promoter in ATSCs were hypomethylated, while in BMSCs they were hypermethylated. The methylation status of Sox9 promoter in BMSCs was only slightly lower than that in ATSCs. Conclusions The epigenetic memory obtained from either bone marrow or adipose tissue favored MSC differentiation along an osteoblastic or adipocytic lineage. The methylation status of the main transcription factors controlling MSC fate contributes to the differential differentiation capacities of different source-derived MSCs. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0716-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Liangliang Xu
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, Special Administrative Region of China
| | - Yamei Liu
- Departments of Diagnostics of Traditional Chinese Medicine, Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, 510006, People's Republic of China
| | - Yuxin Sun
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, Special Administrative Region of China
| | - Bin Wang
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, Special Administrative Region of China
| | - Yunpu Xiong
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, 510240, People's Republic of China
| | - Weiping Lin
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, Special Administrative Region of China
| | - Qiushi Wei
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haibin Wang
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei He
- Key Laboratory of Orthopaedics & Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China. .,Department of Traumatology, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, China.
| | - Bin Wang
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, 510240, People's Republic of China.
| | - Gang Li
- Department of Orthopaedics & Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, Special Administrative Region of China. .,Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, SAR, China. .,Stem Cells and Regenerative Medicine Laboratory, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, Special Administrative Region of China. .,The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, People's Republic of China. .,Room 904, 9/F, Li Ka Shing Institute of Health Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong, Special Administrative Region of China.
| |
Collapse
|
30
|
Crosstalk with Inflammatory Macrophages Shapes the Regulatory Properties of Multipotent Adult Progenitor Cells. Stem Cells Int 2017; 2017:2353240. [PMID: 28785285 PMCID: PMC5529661 DOI: 10.1155/2017/2353240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/27/2017] [Accepted: 06/12/2017] [Indexed: 01/25/2023] Open
Abstract
Macrophages and microglia are key effector cells in immune-mediated neuroinflammatory disorders. Driving myeloid cells towards an anti-inflammatory, tissue repair-promoting phenotype is considered a promising strategy to halt neuroinflammation and promote central nervous system (CNS) repair. In this study, we defined the impact of multipotent adult progenitor cells (MAPC), a stem cell population sharing common mesodermal origin with mesenchymal stem cells (MSCs), on the phenotype of macrophages and the reciprocal interactions between these two cell types. We show that MAPC suppress the secretion of tumor necrosis factor alpha (TNF-α) by inflammatory macrophages partially through a cyclooxygenase 2- (COX-2-) dependent mechanism. In turn, we demonstrate that inflammatory macrophages trigger the immunomodulatory properties of MAPC, including an increased expression of immunomodulatory mediators (e.g., inducible nitric oxide synthase (iNOS) and COX-2), chemokines, and chemokine receptors. Macrophage-primed MAPC secrete soluble factors that suppress TNF-α release by macrophages. Moreover, the MAPC secretome suppresses the antigen-specific proliferation of autoreactive T cells and the T cell stimulatory capacity of macrophages. Finally, MAPC increase their motility towards secreted factors of activated macrophages. Collectively, these in vitro findings reveal intimate reciprocal interactions between MAPC and inflammatory macrophages, which are of importance in the design of MAPC-based therapeutic strategies for neuroinflammatory disorders in which myeloid cells play a crucial role.
Collapse
|
31
|
Li X, Lu X, Sun D, Wang X, Yang L, Zhao S, Nian H, Wei R. Adipose-Derived Mesenchymal Stem Cells Reduce Lymphocytic Infiltration in a Rabbit Model of Induced Autoimmune Dacryoadenitis. Invest Ophthalmol Vis Sci 2017; 57:5161-5170. [PMID: 27699412 PMCID: PMC6016434 DOI: 10.1167/iovs.15-17824] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Purpose To investigate the immunoregulatory roles of adipose-derived mesenchymal stem cells (ADSCs) in autoimmune dacryoadenitis. Methods Rabbits were treated with ADSCs or phosphate-buffered solution on days 1, 3, 5, 7, and 9 after injection of activated peripheral blood lymphocytes, and clinical scores were determined by assessing tear production, break-up time, and fluorescein and hematoxylin and eosin staining. Inflammatory response was determined by measuring the expression of different mediators of inflammation in the lacrimal glands. The Th1/Th17-mediated autoreactive responses were evaluated by determining the proliferative response and the expression of cytokine genes and the lineage-determining transcription factors. The frequency of regulatory T cells (Tregs) was also examined. Results The ADSC-treated rabbits showed decreased autoimmune responses, and the secretory function of their lacrimal gland was restored significantly. Treatment with ADSCs downregulated the Th1 and Th17 responses but enhanced Tregs function. In addition, ADSC treatment noticeably suppressed the expression of matrix metalloproteinase (MMP)-9, MPP-2, IL-1β, and IL-6, whereas it enhanced the expression of the anti-inflammatory cytokine IL-10. Conclusions Our results demonstrated that ADSC administration efficiently ameliorates autoimmune dacryoadenitis mainly via modulating Th1/Th17 responses.
Collapse
Affiliation(s)
- Xue Li
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China
| | - Xiaoxiao Lu
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China
| | - Deming Sun
- Doheny Eye Institute, and Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, California, United States
| | | | - Liyuan Yang
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China
| | - Shaozhen Zhao
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China
| | - Hong Nian
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China
| | - Ruihua Wei
- Tianjin Medical University Eye Hospital, Eye Institute & School of Optometry and Ophthalmology, Tianjin, China
| |
Collapse
|
32
|
Xiao J, Yang R, Biswas S, Zhu Y, Qin X, Zhang M, Zhai L, Luo Y, He X, Mao C, Deng W. Neural Stem Cell-Based Regenerative Approaches for the Treatment of Multiple Sclerosis. Mol Neurobiol 2017; 55:3152-3171. [PMID: 28466274 DOI: 10.1007/s12035-017-0566-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/19/2017] [Indexed: 02/08/2023]
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune, inflammatory, and demyelinating disorder of the central nervous system (CNS), which ultimately leads to axonal loss and permanent neurological disability. Current treatments for MS are largely comprised of medications that are either immunomodulatory or immunosuppressive and are aimed at reducing the frequency and intensity of relapses. Neural stem cells (NSCs) in the adult brain can differentiate into oligodendrocytes in a context-specific manner and are shown to be involved in the remyelination in these patients. NSCs may exert their beneficial effects not only through oligodendrocyte replacement but also by providing trophic support and immunomodulation, a phenomenon now known as "therapeutic plasticity." In this review, we first provided an update on the current knowledge regarding MS pathogenesis and the role of immune cells, microglia, and oligodendrocytes in MS disease progression. Next, we reviewed the current progress on research aimed toward stimulating endogenous NSC proliferation and differentiation to oligodendrocytes in vivo and in animal models of demyelination. In addition, we explored the neuroprotective and immunomodulatory effects of transplanted exogenous NSCs on T cell activation, microglial activation, and endogenous remyelination and their effects on the pathological process and prognosis in animal models of MS. Finally, we examined various protocols to generate genetically engineered NSCs as a potential therapy for MS. Overall, this review highlights the studies involving the immunomodulatory, neurotrophic, and regenerative effects of NSCs and novel methods aiming at stimulating the potential of NSCs for the treatment of MS.
Collapse
Affiliation(s)
- Juan Xiao
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China.,Department of Biological Treatment, Handan Central Hospital, Handan, Hebei, China
| | - Rongbing Yang
- Department of Biological Treatment, Handan Central Hospital, Handan, Hebei, China
| | - Sangita Biswas
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, Guangdong, China. .,Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
| | - Yunhua Zhu
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Xin Qin
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Min Zhang
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Lihong Zhai
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Yi Luo
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Xiaoming He
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Chun Mao
- Department of Neurology, Xiang Yang Central Hospital, Medical College of Hubei University of Arts and Science, Xiangyang, Hubei, China
| | - Wenbin Deng
- School of Pharmaceutical Sciences, Sun Yat-sen University, Shenzhen, Guangdong, China. .,Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
| |
Collapse
|
33
|
Bian P, Ye C, Zheng X, Yang J, Ye W, Wang Y, Zhou Y, Ma H, Han P, Zhang H, Zhang Y, Zhang F, Lei Y, Jia Z. Mesenchymal stem cells alleviate Japanese encephalitis virus-induced neuroinflammation and mortality. Stem Cell Res Ther 2017; 8:38. [PMID: 28209182 PMCID: PMC5314473 DOI: 10.1186/s13287-017-0486-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/30/2016] [Accepted: 01/21/2017] [Indexed: 12/11/2022] Open
Abstract
Background Japanese encephalitis virus (JEV) is the leading cause of viral encephalitis in Asia. Japanese encephalitis (JE) caused by JEV is characterized by extensive inflammatory cytokine secretion, microglia activation, blood-brain barrier (BBB) breakdown, and neuronal death, all of which contribute to the vicious cycle of inflammatory damage. There are currently no effective treatments for JE. Mesenchymal stem cells (MSCs) have been demonstrated to have a therapeutic effect on many central nervous system (CNS) diseases by regulating inflammation and other mechanisms. Methods In vivo, 8- to 10-week-old mice were infected intraperitoneally with JEV and syngeneic bone marrow MSCs were administered through the caudal vein at 1 and 3 days post-infection. The mortality, body weight, and behavior were monitored daily. Brains from each group were harvested at the indicated times for hematoxylin and eosin staining, immunohistochemical observation, flow cytometric analysis, TUNEL staining, Western blot, quantitative real-time polymerase chain reaction, and BBB permeability assays. In vitro, co-culture and mixed culture experiments of MSCs with either microglia or neurons were performed, and then the activation state of microglia and survival rate of neurons were tested 48 h post-infection. Results MSC treatment reduced JEV-induced mortality and improved the recovery from JE in our mouse model. The inflammatory response, microglia activation, neuronal damage, BBB destruction, and viral load (VL) were significantly decreased in the MSC-treated group. In co-culture experiments, MSCs reprogrammed M1-to-M2 switching in microglia and improved neuron survival. Additionally, the VL was decreased in Neuro2a cells in the presence of MSCs accompanied by increased expression of interferon-α/β. Conclusion MSC treatment alleviated JEV-induced inflammation and mortality in mice. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0486-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peiyu Bian
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038, China
| | - Chuantao Ye
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038, China
| | - Xuyang Zheng
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038, China
| | - Jing Yang
- Department of Microbiology, School of Preclinical Medicine, the Fourth Military Medical University, Xi'an, 710032, China
| | - Wei Ye
- Department of Microbiology, School of Preclinical Medicine, the Fourth Military Medical University, Xi'an, 710032, China
| | - Yuan Wang
- Department of Microbiology, School of Preclinical Medicine, the Fourth Military Medical University, Xi'an, 710032, China
| | - Yun Zhou
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038, China
| | - Hongwei Ma
- Department of Microbiology, School of Preclinical Medicine, the Fourth Military Medical University, Xi'an, 710032, China
| | - Peijun Han
- Department of Microbiology, School of Preclinical Medicine, the Fourth Military Medical University, Xi'an, 710032, China
| | - Hai Zhang
- Laboratory Animal Center, the Fourth Military Medical University, Xi'an, 710032, China
| | - Ying Zhang
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038, China
| | - Fanglin Zhang
- Department of Microbiology, School of Preclinical Medicine, the Fourth Military Medical University, Xi'an, 710032, China
| | - Yingfeng Lei
- Department of Microbiology, School of Preclinical Medicine, the Fourth Military Medical University, Xi'an, 710032, China.
| | - Zhansheng Jia
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi'an, 710038, China.
| |
Collapse
|
34
|
Capacity of Human Dental Follicle Cells to Differentiate into Neural Cells In Vitro. Stem Cells Int 2017; 2017:8371326. [PMID: 28261273 PMCID: PMC5316458 DOI: 10.1155/2017/8371326] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 12/28/2016] [Indexed: 01/12/2023] Open
Abstract
The dental follicle is an ectomesenchymal tissue surrounding the developing tooth germ. Human dental follicle cells (hDFCs) have the capacity to commit to differentiation into multiple cell types. Here we investigated the capacity of hDFCs to differentiate into neural cells and the efficiency of a two-step strategy involving floating neurosphere-like bodies for neural differentiation. Undifferentiated hDFCs showed a spindle-like morphology and were positive for neural markers such as nestin, β-III-tubulin, and S100β. The cellular morphology of several cells was neuronal-like including branched dendrite-like processes and neurites. Next, hDFCs were used for neurosphere formation in serum-free medium containing basic fibroblast growth factor, epidermal growth factor, and B27 supplement. The number of cells with neuronal-like morphology and that were strongly positive for neural markers increased with sphere formation. Gene expression of neural markers also increased in hDFCs with sphere formation. Next, gene expression of neural markers was examined in hDFCs during neuronal differentiation after sphere formation. Expression of Musashi-1 and Musashi-2, MAP2, GFAP, MBP, and SOX10 was upregulated in hDFCs undergoing neuronal differentiation via neurospheres, whereas expression of nestin and β-III-tubulin was downregulated. In conclusion, hDFCs may be another optimal source of neural/glial cells for cell-based therapies to treat neurological diseases.
Collapse
|
35
|
Abd Allah SH, Pasha HF, Abdelrahman AA, Mazen NF. Molecular effect of human umbilical cord blood CD34-positive and CD34-negative stem cells and their conjugate in azoospermic mice. Mol Cell Biochem 2017; 428:179-191. [PMID: 28120211 DOI: 10.1007/s11010-016-2928-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Accepted: 12/21/2016] [Indexed: 01/24/2023]
Abstract
Currently, azoospermia is one of the most common diseases of male infertility. Stem cell research is the new hope for novel therapy with a higher degree of safety and lower cost. This study aimed to investigate the effect of umbilical cord blood-derived stem cells (" and mesenchymal "UCB-MSCs") and mono-cell layer implanted into the induced azoospermic mice testis. Stem cells were isolated from umbilical cord blood and CD34+ve cells were separated from negative one by Mini MACs column. At 5th week after single injection of busulfan, stained mesenchymal (CD34-ve), hematopoietic stem cells (CD34+ve) and their conjugate (mono-cell layer) were injected locally into testis. At the end of the study, MSCs group showed that mRNA levels of genes related to meiosis (Vasa, SCP3, and PgK2) were increased with significant decrease of FSH and LH levels, compared to control group. Histologically, most of the tubules restored normal architecture. In contrast, HSCs and mono-cell layer groups showed statically insignificant change of FSH, LH, and gene expression, compared to control group. Histologically, distorted seminiferous tubules, with reduction in sperm content, and interstitial mononuclear cellular infiltration were seen. There was significant increase in the optical density of PCNA immune reaction in MSCs group than azoospermia, HSCs, and mono-cell layer, while there was non-significant difference between MSCs and control group. The present study suggested that injection of MSCs into chemotherapeutic-induced azoospermia in mice improved testicular failure; histologically and functionally, by restoration of spermatogenic gene expression while HSC and mono-cell layer showed no effect on spermatogenesis added to that mono-cell layer may induce testicular tissue damage.
Collapse
Affiliation(s)
- Somia H Abd Allah
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, 2345, Egypt.
| | - Heba F Pasha
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, 2345, Egypt
| | - Abeer A Abdelrahman
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, 2345, Egypt
| | - Nehad F Mazen
- Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
36
|
Kim SY, Kelland EE, Kim JH, Lund BT, Chang X, Wang K, Weiner LP. The influence of retinoic acid on the human oligodendrocyte precursor cells by RNA-sequencing. Biochem Biophys Rep 2016; 9:166-172. [PMID: 29114585 PMCID: PMC5632710 DOI: 10.1016/j.bbrep.2016.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/01/2016] [Accepted: 12/13/2016] [Indexed: 11/16/2022] Open
Abstract
Retinoic acid (RA), a metabolite of vitamin A, has been found to influence regeneration in the adult central nervous system (CNS). There may be an effect of RA in the recovery/repair in multiple sclerosis (MS), an autoimmune and neurodegenerative disease of the CNS. We hypothesized that RA is a regulator of the further differentiation of oligodendrocyte precursor cells (OPCs) – cells key to the remyelination process in MS. We conducted studies utilizing RNA-sequencing in human embryonic stem cell (hESC)-derived neural stem cells (NSCs) and OPCs so as to understand the role of transcriptional regulators during transition from both ESCs to NSCs and NSCs to OPCs. We identified that expression of retinoic acid receptors β and γ (RARβ and RARγ ) was significantly increased following the transition from NSCs to OPCs. We also demonstrated that long term in vitro culture of hESC-derived OPC with different isoforms of RA led to the significant up-regulation of two known transcriptional inhibitors of oligodendrocyte differentiation: Hes5 following prolonged treatment with all-trans-RA, 9-cis RA and 13-cis RA; and Id4 following treatment with 13cisRA. These results suggest that long term exposure to certain RA isoforms may impact the continued differentiation of this population. Retinoic acid (ATRA) might have an effect on generation of oligodendrocyte precursor cells (OPC) in the CNS. RNA-sequencing used for identification of up-regulation of RARβ and RARɣ expression during OPC differentiation. Activation of RARβ and RARɣ by using three different agonists led increase expression of Hes5 and Id4.
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eve E Kelland
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Ji Hong Kim
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brett T Lund
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Xiao Chang
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kai Wang
- Department of Psychiatry and Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Division of Bioinformatics, Department of Preventive medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Leslie P Weiner
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA.,Department of Molecular Microbiology & Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.,Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
37
|
Meamar R, Nematollahi S, Dehghani L, Mirmosayyeb O, Shayegannejad V, Basiri K, Tanhaei AP. The role of stem cell therapy in multiple sclerosis: An overview of the current status of the clinical studies. Adv Biomed Res 2016; 5:46. [PMID: 27110543 PMCID: PMC4817403 DOI: 10.4103/2277-9175.178791] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 08/19/2014] [Indexed: 01/01/2023] Open
Abstract
The complexity of multiple sclerosis (MS) and the incompetence of a large number of promised treatments for MS urge us to plan new and more effective therapeutic approaches that aim to suppress ongoing autoimmune responses and induction of local endogenous regeneration. Emerging data propose that hematopoietic, mesenchymal, and neural stem cells have the potential to restore self-tolerance, provide in situ immunomodulation and neuroprotection, as well as promote regeneration. Thus, in this article, we will first provide an overview of the cell sources for proposed mechanisms that contribute to the beneficial effects of stem cell transplantation, the ideal route and/or timing of stem cell-based therapies for each main stem cell group, and finally, an overview of the current status of stem cell research in clinical trial stages in MS by comparable and healthy therapeutic effects of different stem cell therapies for MS patients.
Collapse
Affiliation(s)
- Rokhsareh Meamar
- Department of Medical Sciences, Islamic Azad University, Najafabad Branch, Tehran, Iran
- Isfahan Neurosciences Research Center, Al Zahra Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrzad Nematollahi
- PhD Candidate in Epidemiology, School of Public Health and Institute of Public Health Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Dehghani
- Department of Medical Sciences, Islamic Azad University, Najafabad Branch, Tehran, Iran
- Isfahan Neurosciences Research Center, Al Zahra Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Mirmosayyeb
- Isfahan Neurosciences Research Center, Al Zahra Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Shayegannejad
- Isfahan Neurosciences Research Center, Al Zahra Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Keivan Basiri
- Isfahan Neurosciences Research Center, Al Zahra Hospital, Tehran University of Medical Sciences, Tehran, Iran
- Department of Neurology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amir Pouya Tanhaei
- Isfahan Neurosciences Research Center, Al Zahra Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Ma Y, Wang J, Wang Y, Yang GY. The biphasic function of microglia in ischemic stroke. Prog Neurobiol 2016; 157:247-272. [PMID: 26851161 DOI: 10.1016/j.pneurobio.2016.01.005] [Citation(s) in RCA: 505] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 12/22/2015] [Accepted: 01/10/2016] [Indexed: 12/16/2022]
Abstract
Microglia are brain resident macrophages originated from primitive progenitor cells in the yolk sac. Microglia can be activated within hours and recruited to the lesion site. Traditionally, microglia activation is considered to play a deleterious role in ischemic stroke, as inhibition of microglia activation attenuates ischemia induced brain injury. However, increasing evidence show that microglia activation is critical for attenuating neuronal apoptosis, enhancing neurogenesis, and promoting functional recovery after cerebral ischemia. Differential polarization of microglia could likely explain the biphasic role of microglia in ischemia. We comprehensively reviewed the mechanisms involved in regulating microglia activation and polarization. The latest discoveries of microRNAs in modulating microglia function are discussed. In addition, the interaction between microglia and other cells including neurons, astrocytes, oligodendrocytes, and stem cells were also reviewed. Future therapies targeting microglia may not exclusively aim at suppressing microglia activation, but also at modulating microglia polarization at different stages of ischemic stroke. More work is needed to elucidate the cellular and molecular mechanisms of microglia polarization under ischemic environment. The roles of microRNAs and transplanted stem cells in mediating microglia activation and polarization during brain ischemia also need to be further studied.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jixian Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; Department of Rehabilitation, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
39
|
Trubiani O, Giacoppo S, Ballerini P, Diomede F, Piattelli A, Bramanti P, Mazzon E. Alternative source of stem cells derived from human periodontal ligament: a new treatment for experimental autoimmune encephalomyelitis. Stem Cell Res Ther 2016; 7:1. [PMID: 26729060 PMCID: PMC4700621 DOI: 10.1186/s13287-015-0253-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/15/2015] [Accepted: 12/02/2015] [Indexed: 01/08/2023] Open
Abstract
Background Multiple sclerosis is a demyelinating disease mostly of autoimmune origin that affects and damages the central nervous system, leading to a disabling condition. The aim of the present study was to investigate whether administration of mesenchymal stem cells from human periodontal ligament (hPDLSCs) could ameliorate multiple sclerosis progression by exerting neuroprotective effects in an experimental model of autoimmune encephalomyelitis (EAE). Methods EAE was induced by immunization with myelin oligodendroglial glycoprotein peptide (MOG)35–55 in C57BL/6 mice. After immunization, mice were observed every 48 hours for signs of EAE and weight loss. At the onset of disease, approximately 14 days after immunization, EAE mice were subjected to a single intravenous injection of hPDLSCs (106 cells/150 μl) into the tail vein. At the point of animal sacrifice on day 56 after EAE induction, spinal cord and brain tissues were collected in order to perform histological evaluation, immunohistochemistry and western blotting analysis. Results Achieved results reveal that treatment with hPDLSCs may exert neuroprotective effects against EAE, diminishing both clinical signs and histological score typical of the disease (lymphocytic infiltration and demyelination) probably through the production of neurotrophic factors (results focused on brain-derived neurotrophic factor and nerve growth factor expression). Furthermore, administration of hPDLSCs modulates expression of inflammatory key markers (tumor necrosis factor-α, interleukin (IL)-1β, IL-10, glial fibrillary acidic protein, Nrf2 and Foxp3), the release of CD4 and CD8α T cells, and the triggering of apoptotic death pathway (data shown for cleaved caspase 3, p53 and p21). Conclusions In light of the achieved results, transplantation of hPDLSCs may represent a putative novel and helpful tool for multiple sclerosis treatment. These cells could have considerable implication for future therapies for multiple sclerosis and this study may represent the starting point for further investigations.
Collapse
Affiliation(s)
- Oriana Trubiani
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti-Pescara, via dei Vestini, 31, 66100, Chieti, Italy.
| | - Sabrina Giacoppo
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, contrada Casazza, 98124, Messina, Italy.
| | - Patrizia Ballerini
- Department of Psychological, Humanities and Territorial Sciences, University "G. d'Annunzio" Chieti-Pescara, Chieti, Italy.
| | - Francesca Diomede
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti-Pescara, via dei Vestini, 31, 66100, Chieti, Italy.
| | - Adriano Piattelli
- Stem Cells and Regenerative Medicine Laboratory, Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio", Chieti-Pescara, via dei Vestini, 31, 66100, Chieti, Italy.
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, contrada Casazza, 98124, Messina, Italy.
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, contrada Casazza, 98124, Messina, Italy.
| |
Collapse
|
40
|
Scutellarin Alleviates Behavioral Deficits in a Mouse Model of Multiple Sclerosis, Possibly Through Protecting Neural Stem Cells. J Mol Neurosci 2015; 58:210-20. [PMID: 26514969 DOI: 10.1007/s12031-015-0660-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/24/2015] [Indexed: 12/14/2022]
Abstract
Scutellarin, a flavonoid extracted from an herbal medication (Erigeron breviscapus Hand-Mazz), has been shown to protect neurons against damage and to promote neurogenesis, and thus has therapeutic potential in the treatment of a variety of neurodegenerative diseases. Since neural stem cells (NSCs) could differentiate into myelin-producing oligodendrocytes, we speculate that scutellarin could also be used to treat multiple sclerosis (MS). In the current study, we examined potential effects of scutellarin using a mouse model of MS. Briefly, adult C57BL/6 mice exposed to cuprizone (8 mg/day through diet, for 6 consecutive weeks) randomly received scutellarin (50 mg/kg/day) or vehicle for 10 consecutive days. In the scutellarin-treated group, rotarod testing at the end of the treatment showed significant improvement of motor function (increased time to fall); myelin basic protein (MBP) staining of the corpus callosum revealed decreased demyelination; TUNEL staining followed by Nestin or Sox2 staining revealed increased number of NSCs and decreased rate of NSC apoptosis in the subventricular zone (SVZ) of the lateral ventricles (LV). In a series of experiments using cultured NSCs subjected to cuprizone injury, we confirmed the protective effects of scutellarin. At 30 μM, scutellarin increased the commitment of NSCs to the oligodendrocyte and neuronal lineages, as evidenced by NG2 chondroitin sulfate proteoglycan (NG2) and doublecortin (DCX) staining. Differentiation into astrocytes (as revealed by glial fibrillary acidic protein (GFAP) staining) was decreased. Maturation of the NSCs committed to the oligodendrocyte lineage, as evidenced by oligodendrocyte marker O4 antibody (O4) staining and MBP staining, was also promoted by scutellarin. Further analysis revealed that scutellarin might suppress the phosphorylation of p38 in cuprizone-induced NSCs. In summary, scutellarin could alleviate motor deficits in a mouse model for MS, possibly by inhibiting NSC apoptosis and promoting differentiation of NSCs to myelin-producing oligodendrocytes.
Collapse
|
41
|
Li T, Li Z, Nan F, Dong J, Deng Y, Yu Q, Zhang T. Construction of a novel inducing system with multi-layered alginate microcapsules to regulate differentiation of neural precursor cells from bone mesenchymal stem cells. Med Hypotheses 2015; 85:910-3. [PMID: 26386487 DOI: 10.1016/j.mehy.2015.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 09/09/2015] [Indexed: 01/08/2023]
Abstract
Neural precursor cells (NPCs) are a promising cell source for the treatment of nervous system diseases; however, they are limited in their applications due to source-related ethical considerations or legislations. Therefore, a novel approach is necessary to obtain sufficient NPCs. Recently, the usage of bone marrow-derived mesenchymal stem cells (BMSCs) differentiated into neural cells has become a potential method to obtain NPCs. Moreover, growth factors (GFs) are emerging as inducers to evoke the differentiation of BMSCs into NPCs. For example, GFs may activate various signaling pathways related to neural differentiation, such as phosphatidylinositol 3 kinase/protein kinase B, cyclic adenosine monophosphate/protein kinase A, and Janus kinase/signal transducer activator of transcription. However, the utilization of growth factors still has some limitations such as high costs and low rates of neural differentiation. Neuroblastoma cells have been characterized as a potential pool for growth factors. Additionally, basic fibroblast growth factor (bFGF), a type of growth factor, has been demonstrated to be able to increase the differentiation and survival rate of NPCs. For better use of neuroblastoma cells and bFGF, we established a novel system involving multi-layered alginate-polylysine-alginate (APA) microcapsules to encapsulate neuroblastoma cells and bFGF, which may not only provide sufficient growth factors in a sustained manner but also avoid the carcinogenicity caused by neuroblastoma cells. Above all, we hypothesized that neuroblastoma cells and bFGF encapsulated in multilayered alginate microcapsules may efficiently induce the differentiation of BMSCs into NPCs.
Collapse
Affiliation(s)
- Tao Li
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Zhengwei Li
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Feng Nan
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China.
| | - Jianli Dong
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Yushuang Deng
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Qing Yu
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| | - Teng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, District Shahekou, Dalian 116023, PR China
| |
Collapse
|
42
|
Mozafari S, Laterza C, Roussel D, Bachelin C, Marteyn A, Deboux C, Martino G, Baron-Van Evercooren A. Skin-derived neural precursors competitively generate functional myelin in adult demyelinated mice. J Clin Invest 2015; 125:3642-56. [PMID: 26301815 DOI: 10.1172/jci80437] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/07/2015] [Indexed: 12/26/2022] Open
Abstract
Induced pluripotent stem cell-derived (iPS-derived) neural precursor cells may represent the ideal autologous cell source for cell-based therapy to promote remyelination and neuroprotection in myelin diseases. So far, the therapeutic potential of reprogrammed cells has been evaluated in neonatal demyelinating models. However, the repair efficacy and safety of these cells has not been well addressed in the demyelinated adult CNS, which has decreased cell plasticity and scarring. Moreover, it is not clear if these induced pluripotent-derived cells have the same reparative capacity as physiologically committed CNS-derived precursors. Here, we performed a side-by-side comparison of CNS-derived and skin-derived neural precursors in culture and following engraftment in murine models of adult spinal cord demyelination. Grafted induced neural precursors exhibited a high capacity for survival, safe integration, migration, and timely differentiation into mature bona fide oligodendrocytes. Moreover, grafted skin-derived neural precursors generated compact myelin around host axons and restored nodes of Ranvier and conduction velocity as efficiently as CNS-derived precursors while outcompeting endogenous cells. Together, these results provide important insights into the biology of reprogrammed cells in adult demyelinating conditions and support use of these cells for regenerative biomedicine of myelin diseases that affect the adult CNS.
Collapse
|
43
|
Yang RF, Liu TH, Zhao K, Xiong CL. Enhancement of mouse germ cell-associated genes expression by injection of human umbilical cord mesenchymal stem cells into the testis of chemical-induced azoospermic mice. Asian J Androl 2015; 16:698-704. [PMID: 24830694 PMCID: PMC4215652 DOI: 10.4103/1008-682x.129209] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Various methods are currently under investigation to preserve fertility in males treated with high-dose chemotherapy and radiation for malignant and nonmalignant disorders. Human umbilical cord mesenchymal stem cells (HUC-MSCs), which possess potent immunosuppressive function and secrete various cytokines and growth factors, have the potential clinical applications. As a potential alternative, we investigate whether injection of HUC-MSCs into the interstitial compartment of the testes to promote spermatogenic regeneration efficiently. HUC-MSCs were isolated from different sources of umbilical cords and injected into the interstitial space of one testis from 10 busulfan-treated mice (saline and HEK293 cells injections were performed in a separate set of mice) and the other testis remained uninjected. Three weeks after MSCs injection, Relative quantitative reverse transcription polymerase chain reaction was used to identify the expression of 10 of germ cell associated, which are all related to meiosis, demonstrated higher levels of spermatogenic gene expression (2–8 fold) in HUC-MSCs injected testes compared to the contralateral uninjected testes (five mice). Protein levels for germ cell-specific genes, miwi, vasa and synaptonemal complex protein (Scp3) were also higher in MSC-treated testes compared to injected controls 3 weeks after treatment. However, no different expression was detected in saline water and HEK293 cells injection control group. We have demonstrated HUC-MSCs could affect mouse germ cell-specific genes expression. The results also provide a possibility that the transplanted HUC-MSCs may promote the recovery of spermatogenesis. This study provides further evidence for preclinical therapeutic effects of HUC-MSCs, and explores a new approach to the treatment of azoospermia.
Collapse
Affiliation(s)
| | | | | | - Cheng-Liang Xiong
- Center of Reproductive Medicine, Family Planning Research Institute, Tongji Medical College, Huazhong University of Science and Technology; Wuhan Tongji Reproductive Medicine Hospital, Wuhan, China
| |
Collapse
|
44
|
Weinger JG, Plaisted WC, Maciejewski SM, Lanier LL, Walsh CM, Lane TE. Activating receptor NKG2D targets RAE-1-expressing allogeneic neural precursor cells in a viral model of multiple sclerosis. Stem Cells 2015; 32:2690-701. [PMID: 24898518 DOI: 10.1002/stem.1760] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 04/25/2014] [Accepted: 04/25/2014] [Indexed: 12/11/2022]
Abstract
Transplantation of major histocompatibility complex-mismatched mouse neural precursor cells (NPCs) into mice persistently infected with the neurotropic JHM strain of mouse hepatitis virus (JHMV) results in rapid rejection that is mediated, in part, by T cells. However, the contribution of the innate immune response to allograft rejection in a model of viral-induced neurological disease has not been well defined. Herein, we demonstrate that the natural killer (NK) cell-expressing-activating receptor NKG2D participates in transplanted allogeneic NPC rejection in mice persistently infected with JHMV. Cultured NPCs derived from C57BL/6 (H-2(b) ) mice express the NKG2D ligand retinoic acid early precursor transcript (RAE)-1 but expression was dramatically reduced upon differentiation into either glia or neurons. RAE-1(+) NPCs were susceptible to NK cell-mediated killing whereas RAE-1(-) cells were resistant to lysis. Transplantation of C57BL/6-derived NPCs into JHMV-infected BALB/c (H-2(d) ) mice resulted in infiltration of NKG2D(+) CD49b(+) NK cells and treatment with blocking antibody specific for NKG2D increased survival of allogeneic NPCs. Furthermore, transplantation of differentiated RAE-1(-) allogeneic NPCs into JHMV-infected BALB/c mice resulted in enhanced survival, highlighting a role for the NKG2D/RAE-1 signaling axis in allograft rejection. We also demonstrate that transplantation of allogeneic NPCs into JHMV-infected mice resulted in infection of the transplanted cells suggesting that these cells may be targets for infection. Viral infection of cultured cells increased RAE-1 expression, resulting in enhanced NK cell-mediated killing through NKG2D recognition. Collectively, these results show that in a viral-induced demyelination model, NK cells contribute to rejection of allogeneic NPCs through an NKG2D signaling pathway.
Collapse
Affiliation(s)
- Jason G Weinger
- Department of Molecular Biology & Biochemistry; Sue and Bill Gross Stem Cell Center, University of California, Irvine, California, USA
| | | | | | | | | | | |
Collapse
|
45
|
Burns TC, Verfaillie CM. From mice to mind: Strategies and progress in translating neuroregeneration. Eur J Pharmacol 2015; 759:90-100. [PMID: 25814255 DOI: 10.1016/j.ejphar.2015.03.041] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/18/2015] [Accepted: 03/12/2015] [Indexed: 12/20/2022]
Abstract
Decisions about what experimental therapies are advanced to clinical trials are based almost exclusively on findings in preclinical animal studies. Over the past 30 years, animal models have forecast the success of hundreds of neuroprotective pharmacological therapies for stroke, Alzheimer׳s disease, spinal cord injury, traumatic brain injury and amyotrophic lateral sclerosis. Yet almost without exception, all have failed. Rapid advances in stem cell technologies have raised new hopes that these neurological diseases may one day be treatable. Still, how can neuroregenerative therapies be translated into clinical realities if available animal models are such poor surrogates of human disease? To address this question we discuss human and rodent neurogenesis, evaluate mechanisms of action for cellular therapies and describe progress in translating neuroregeneration to date. We conclude that not only are appropriate animal models critical to the development of safe and effective therapies, but that the multiple mechanisms of stem cell-mediated therapies may be particularly well suited to the mechanistically diverse nature of central nervous system diseases in mice and man.
Collapse
Affiliation(s)
- Terry C Burns
- Department of Neurosurgery and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, USA.
| | | |
Collapse
|
46
|
Marrelli M, Paduano F, Tatullo M. Human periapical cyst-mesenchymal stem cells differentiate into neuronal cells. J Dent Res 2015; 94:843-52. [PMID: 25672890 DOI: 10.1177/0022034515570316] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
It was recently reported that human periapical cysts (hPCys), a commonly occurring odontogenic cystic lesion of inflammatory origin, contain mesenchymal stem cells (MSCs) with the capacity for self-renewal and multilineage differentiation. In this study, periapical inflammatory cysts were compared with dental pulp to determine whether this tissue may be an alternative accessible tissue source of MSCs that retain the potential for neurogenic differentiation. Flow cytometry and immunofluorescence analysis indicated that hPCy-MSCs and dental pulp stem cells spontaneously expressed the neuron-specific protein β-III tubulin and the neural stem-/astrocyte-specific protein glial fibrillary acidic protein (GFAP) in their basal state before differentiation occurs. Furthermore, undifferentiated hPCy-MSCs showed a higher expression of transcripts for neuronal markers (β-III tubulin, NF-M, MAP2) and neural-related transcription factors (MSX-1, Foxa2, En-1) as compared with dental pulp stem cells. After exposure to neurogenic differentiation conditions (neural media containing epidermal growth factor [EGF], basic fibroblast growth factor [bFGF], and retinoic acid), the hPCy-MSCs showed enhanced expression of β-III tubulin and GFAP proteins, as well as increased expression of neurofilaments medium, neurofilaments heavy, and neuron-specific enolase at the transcript level. In addition, neurally differentiated hPCy-MSCs showed upregulated expression of the neural transcription factors Pitx3, Foxa2, Nurr1, and the dopamine-related genes tyrosine hydroxylase and dopamine transporter. The present study demonstrated for the first time that hPCy-MSCs have a predisposition toward the neural phenotype that is increased when exposed to neural differentiation cues, based on upregulation of a comprehensive set of proteins and genes that define neuronal cells. In conclusion, these results provide evidence that hPCy-MSCs might be another optimal source of neural/glial cells for cell-based therapies to treat neurologic diseases.
Collapse
Affiliation(s)
- M Marrelli
- Unit of Maxillofacial Surgery, Calabrodental, Crotone, Italy
| | - F Paduano
- Tecnologica Research Institute, Biomedical Section, Crotone, Italy
| | - M Tatullo
- Tecnologica Research Institute, Biomedical Section, Crotone, Italy
| |
Collapse
|
47
|
Michailidou I, de Vries HE, Hol EM, van Strien ME. Activation of endogenous neural stem cells for multiple sclerosis therapy. Front Neurosci 2015; 8:454. [PMID: 25653584 PMCID: PMC4299409 DOI: 10.3389/fnins.2014.00454] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 12/22/2014] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disorder of the central nervous system, leading to severe neurological deficits. Current MS treatment regimens, consist of immunomodulatory agents aiming to reduce the rate of relapses. However, these agents are usually insufficient to treat chronic neurological disability. A promising perspective for future therapy of MS is the regeneration of lesions with replacement of the damaged oligodendrocytes or neurons. Therapies targeting to the enhancement of endogenous remyelination, aim to promote the activation of either the parenchymal oligodendrocyte progenitor cells or the subventricular zone-derived neural stem cells (NSCs). Less studied but highly potent, is the strategy of neuronal regeneration with endogenous NSCs that although being linked to numerous limitations, is anticipated to ameliorate cognitive disability in MS. Focusing on the forebrain, this review highlights the role of NSCs in the regeneration of MS lesions.
Collapse
Affiliation(s)
- Iliana Michailidou
- Department of Astrocyte Biology and Neurodegeneration, The Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Sciences Amsterdam, Netherlands
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, VU University Medical Center Amsterdam, Netherlands
| | - Elly M Hol
- Department of Astrocyte Biology and Neurodegeneration, The Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Sciences Amsterdam, Netherlands ; Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam Amsterdam, Netherlands ; Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Miriam E van Strien
- Department of Astrocyte Biology and Neurodegeneration, The Netherlands Institute for Neuroscience, An Institute of the Royal Netherlands Academy of Sciences Amsterdam, Netherlands ; Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| |
Collapse
|
48
|
Czepiel M, Boddeke E, Copray S. Human oligodendrocytes in remyelination research. Glia 2014; 63:513-30. [DOI: 10.1002/glia.22769] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 11/05/2014] [Indexed: 01/04/2023]
Affiliation(s)
- Marcin Czepiel
- Department of Neuroscience; University Medical Center Groningen; A.Deusinglaan 1, 9713AV Groningen The Netherlands
| | - Erik Boddeke
- Department of Neuroscience; University Medical Center Groningen; A.Deusinglaan 1, 9713AV Groningen The Netherlands
| | - Sjef Copray
- Department of Neuroscience; University Medical Center Groningen; A.Deusinglaan 1, 9713AV Groningen The Netherlands
| |
Collapse
|
49
|
Donders R, Vanheusden M, Bogie JFJ, Ravanidis S, Thewissen K, Stinissen P, Gyselaers W, Hendriks JJA, Hellings N. Human Wharton's Jelly-Derived Stem Cells Display Immunomodulatory Properties and Transiently Improve Rat Experimental Autoimmune Encephalomyelitis. Cell Transplant 2014; 24:2077-98. [PMID: 25310756 DOI: 10.3727/096368914x685104] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Umbilical cord matrix or Wharton's jelly-derived stromal cells (WJ-MSCs) are an easily accessible source of mesenchymal-like stem cells. Recent studies describe a hypoimmunogenic phenotype, multipotent differentiation potential, and trophic support function for WJ-MSCs, with variable clinical benefit in degenerative disease models such as stroke, myocardial infarction, and Parkinson's disease. It remains unclear whether WJ-MSCs have therapeutic value for multiple sclerosis (MS), where autoimmune-mediated demyelination and neurodegeneration need to be halted. In this study, we investigated whether WJ-MSCs possess the required properties to effectively and durably reverse these pathological hallmarks and whether they survive in an inflammatory environment after transplantation. WJ-MSCs displayed a lowly immunogenic phenotype and showed intrinsic expression of neurotrophic factors and a variety of anti-inflammatory molecules. Furthermore, they dose-dependently suppressed proliferation of activated T cells using contact-dependent and paracrine mechanisms. Indoleamine 2,3-dioxygenase 1 was identified as one of the main effector molecules responsible for the observed T-cell suppression. The immune-modulatory phenotype of WJ-MSCs was further enhanced after proinflammatory cytokine treatment in vitro (licensing). In addition to their effect on adaptive immunity, WJ-MSCs interfered with dendritic cell differentiation and maturation, thus directly affecting antigen presentation and therefore T-cell priming. Systemically infused WJ-MSCs potently but transiently ameliorated experimental autoimmune encephalomyelitis (EAE), an animal model for MS, when injected at onset or during chronic disease. This protective effect was paralleled with a reduction in autoantigen-induced T-cell proliferation, confirming their immunomodulatory activity in vivo. Surprisingly, in vitro licensed WJ-MSCs did not ameliorate EAE, indicative of a fast rejection as a result of enhanced immunogenicity. Collectively, we show that WJ-MSCs have trophic support properties and effectively modulate immune cell functioning both in vitro and in the EAE model, suggesting WJ-MSC may hold promise for MS therapy. Future research is needed to optimize survival of stem cells and enhance clinical durability.
Collapse
Affiliation(s)
- Raf Donders
- Hasselt University, Biomedical Research Institute/Transnational University Limburg, School of Life Sciences, Diepenbeek, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lublin FD, Bowen JD, Huddlestone J, Kremenchutzky M, Carpenter A, Corboy JR, Freedman MS, Krupp L, Paulo C, Hariri RJ, Fischkoff SA. Human placenta-derived cells (PDA-001) for the treatment of adults with multiple sclerosis: a randomized, placebo-controlled, multiple-dose study. Mult Scler Relat Disord 2014; 3:696-704. [PMID: 25891548 DOI: 10.1016/j.msard.2014.08.002] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 07/02/2014] [Accepted: 08/14/2014] [Indexed: 01/21/2023]
Abstract
BACKGROUND Infusion of PDA-001, a preparation of mesenchymal-like cells derived from full-term human placenta, is a new approach in the treatment of patients with multiple sclerosis. OBJECTIVE This safety study aimed to rule out the possibility of paradoxical exacerbation of disease activity by PDA-001 in patients with multiple sclerosis. METHODS This was a phase 1b, multicenter, randomized, double-blind, placebo-controlled, 2-dose ranging study including patients with relapsing-remitting multiple sclerosis or secondary progressive multiple sclerosis. The study was conducted at 6 sites in the United States and 2 sites in Canada. Patients were randomized 3:1 to receive 2 low-dose infusions of PDA-001 (150×10(6) cells) or placebo, given 1 week apart. After completing this cohort, subsequent patients received high-dose PDA-001 (600×10(6) cells) or placebo. Monthly brain magnetic resonance imaging scans were performed. The primary end point was ruling out the possibility of paradoxical worsening of MS disease activity. This was monitored using Cutter׳s rule (≥5 new gadolinium lesions on 2 consecutive scans) by brain magnetic resonance imaging on a monthly basis for six months and also the frequency of multiple sclerosis relapse. RESULTS Ten patients with relapsing-remitting multiple sclerosis and 6 with secondary progressive multiple sclerosis were randomly assigned to treatment: 6 to low-dose PDA-001, 6 to high-dose PDA-001, and 4 to placebo. No patient met Cutter׳s rule. One patient receiving high-dose PDA-001 had an increase in T2 and gadolinium lesions and in Expanded Disability Status Scale score during a multiple sclerosis flare 5 months after receiving PDA-001. No other patient had an increase in Expanded Disability Status Scale score>0.5, and most had stable or decreasing Expanded Disability Status Scale scores. With high-dose PDA-001, 1 patient experienced a grade 1 anaphylactoid reaction and 1 had grade 2 superficial thrombophlebitis. Other adverse events were mild to moderate and included headache, fatigue, infusion site reactions, and urinary tract infection. CONCLUSION PDA-001 infusions were safe and well tolerated in relapsing-remitting multiple sclerosis and secondary progressive multiple sclerosis patients. No paradoxical worsening of lesion counts was noted with either dose.
Collapse
Affiliation(s)
- Fred D Lublin
- Icahn School of Medicine at Mount Sinai, 5 East 98th Street, Box 1138, New York, NY 10029, United States.
| | - James D Bowen
- Swedish Neuroscience Institute, 1600 East Jefferson, Suite 205, Seattle, WA 98122, United States.
| | - John Huddlestone
- MultiCare Health System-Neuroscience Center of Washington, 915 6th Avenue, Suite 101 and 200, Tacoma, WA 98405, United States.
| | - Marcelo Kremenchutzky
- London Health Sciences Centre, University Hospital, Multiple Sclerosis Clinic, 7th Floor, 339 Windermere Road, PO Box 5339, London, ON, Canada N6A 5A5.
| | - Adam Carpenter
- University of Minnesota, Clinical Neuroscience Research Unit, 717 Delaware Street SE, Suite 246, Minneapolis, MN 55414, United States.
| | - John R Corboy
- University of Colorado Denver, 12631 E. 17th Avenue, Mail Stop B185, Room 5506, Aurora, CO 80045, United States.
| | - Mark S Freedman
- The Ottawa Hospital Multiple Sclerosis Clinic, 501 Smyth Road, Box 607, Ottawa, Ontario, Canada K1H 8L6.
| | - Lauren Krupp
- MS Comprehensive Care Center, MSC T12-020 SUNY, Stony Brook, NY 11794, United States.
| | - Corri Paulo
- Celgene Cellular Therapeutics, 7 Powderhorn Road, Warren, NJ 07059, United States.
| | - Robert J Hariri
- Celgene Cellular Therapeutics, 7 Powderhorn Road, Warren, NJ 07059, United States.
| | - Steven A Fischkoff
- Celgene Cellular Therapeutics, 7 Powderhorn Road, Warren, NJ 07059, United States.
| |
Collapse
|