1
|
Lotfy A, Ali NS, Abdelgawad M, Salama M. Mesenchymal stem cells as a treatment for multiple sclerosis: a focus on experimental animal studies. Rev Neurosci 2021; 31:161-179. [PMID: 31605598 DOI: 10.1515/revneuro-2019-0040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is a progressive and debilitating neurological condition in which the immune system abnormally attacks the myelin sheath insulating the nerves. Mesenchymal stem cells (MSCs) are found in most adult tissues and play a significant systemic role in self-repair. MSCs have promising therapeutic effects in many diseases, such as autoimmune diseases, including MS. MSCs have been tested in MS animal models, such as experimental autoimmune encephalomyelitis. Other studies have combined other agents with MSCs, genetically modified MSCs, or used culture medium from MSCs. In this review, we will summarize these studies and compare the main factors in each study, such as the source of MSCs, the type of animal model, the route of injection, the number of injected cells, and the mechanism of action.
Collapse
Affiliation(s)
- Ahmed Lotfy
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt, e-mail:
| | - Nourhan S Ali
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt
| | - Mai Abdelgawad
- Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt
| | - Mohamed Salama
- Medical Experimental Research Center (MERC), Faculty of Medicine, Mansoura University, Mansourah, Ad Daqahliyah, Egypt.,Institute of Global Health and Human Ecology (IGHHE), American University in Cairo (AUC), Cairo, Egypt
| |
Collapse
|
2
|
Tahmasebi F, Pasbakhsh P, Barati S, Madadi S, Kashani IR. The effect of microglial ablation and mesenchymal stem cell transplantation on a cuprizone-induced demyelination model. J Cell Physiol 2021; 236:3552-3564. [PMID: 32996165 DOI: 10.1002/jcp.30090] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is a demyelinating autoimmune disease of the central nervous system with symptoms such as neuroinflammation, astrocytosis, microgliosis, and axonal degeneration. Mesenchymal stem cells (MSCs) with their immunomodulation, differentiation, and neuroprotection abilities can influence the remyelination process. The goal of this study is to investigate the impact of microglial ablation and MSCs transplantation on remyelination processes in the corpus callosum (CC) of the cuprizone demyelination model. For the induction of a chronic demyelination model, C57BL6 mice were fed with chow containing 0.2% cuprizone (wt/wt) for 12 weeks. For the depletion of microglia, PLX3397 was used as a colony-stimulating factor 1 receptor inhibitor for 21 days. MSCs were injected to the right lateral ventricle and after 2 weeks, the mice were killed. We assessed glial cells using specific markers such as APC, Iba-1, and GFAP using the immunohistochemistry method. Remyelination was evaluated by Luxol fast blue (LFB) staining and transmission electron microscope (TEM). The specific genes of microglia and MSCs were evaluated by a quantitative real-time polymerase chain reaction. According to the results of the study, 21 days of PLX3397 treatment significantly reduced microglial cells, and MSCs transplantation decreased the number of astrocytes, whereas the oligodendrocytes population increased significantly in PLX + MSC group in comparison with the cuprizone mice. Furthermore, PLX and MSC treatment elevated levels of remyelination compared with the cuprizone group, as confirmed by LFB staining and TEM analysis. The molecular results showed that MSC transplantation significantly decreased the number of microglia through the CX3CL1/CX3CR1 axis. These results revealed that PLX3397 treatment and MSCs injection reduced microgliosis and astrocytosis. It also increased the oligodendrocytes population by enhancing remyelination in the CC of the cuprizone model of MS.
Collapse
Affiliation(s)
- Fatemeh Tahmasebi
- Department of Anatomy, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Barati
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soheila Madadi
- Department of Anatomy, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Iraj R Kashani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
3
|
Hu R, Lv W, Zhang S, Liu Y, Sun B, Meng Y, Kong Q, Mu L, Wang G, Zhang Y, Li H, Liu X. Combining miR-23b exposure with mesenchymal stem cell transplantation enhances therapeutic effects on EAE. Immunol Lett 2020; 229:18-26. [PMID: 33238163 DOI: 10.1016/j.imlet.2020.11.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/01/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023]
Abstract
Bone marrow mesenchymal stem cells (BMSCs) have the immuno-modulatory capacity to ameliorate autoimmune diseases, such as multiple schlerosis (MS), systemic lupus erythematosus and rheumatoid arthritis. However, BMSC-mediated immunosuppression can be challenging to achieve. The efficacy of BMSC transplantation may be augmented by an adjuvant therapy. Here, we demonstrated that treatment of mice with experimental autoimmune encephalomyelitis (EAE), a model of MS, with BMSCs over-expressing microRNA (miR)-23b provided better synergistic and longer-term therapeutic effects than treatment with traditional BMSCs. Over-expression of miR-23b enhanced the ability of BMSCs to inhibit differentiation of Th17 cells and reduced IL-17 secretion. Compared to traditional BMSCs, the miR-23b over-expressing BMSCs (miR23b-BMSCs) exhibited enhanced secretion of tumor growth factor beta 1 (TGF-β1), a cytokine that promotes the differentiation of regulatory T (Treg) cells. Pathologically, miR23b-BMSC transplantation delayed EAE progression, apparently by reducing the Th17/Treg cell ratio and inhibiting inflammatory cell infiltration across the blood-brain barrier, and thus slowing spinal cord demyelination. These results may lead to better utility of BMSCs as a treatment for autoimmune diseases.
Collapse
Affiliation(s)
- Ruixue Hu
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Weiqi Lv
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Sifan Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yumei Liu
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Bo Sun
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yanting Meng
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Lili Mu
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Guangyou Wang
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yao Zhang
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xijun Liu
- Department of Neurobiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
4
|
O'Sullivan A, Lange S, Rotheneichner P, Bieler L, Aigner L, Rivera FJ, Couillard-Despres S. Dimethylsulfoxide Inhibits Oligodendrocyte Fate Choice of Adult Neural Stem and Progenitor Cells. Front Neurosci 2019; 13:1242. [PMID: 31849577 PMCID: PMC6901908 DOI: 10.3389/fnins.2019.01242] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 11/04/2019] [Indexed: 12/13/2022] Open
Abstract
Several clinical trials address demyelinating diseases via transplantation of mesenchymal stromal cells (MSCs). Published reports detail that administration of MSCs in patients may provide a beneficial immunomodulation, and that factors secreted by MSCs are potent inducers of oligodendrogenesis. Dimethylsulfoxide (DMSO) is widely used in life science and medicine as solvent, vehicle or cryoprotectant for cells used in transplantation. Importantly, most transplantation protocols do not include the removal of DMSO before injecting the cell suspension into patients. This indifferent application of DMSO is coming under increasing scrutiny following reports investigating its potential toxic side-effects. While the impact of DMSO on the central nervous system (CNS) has been partially studied, its effect on oligodendrocytes and oligodendrogenesis has not been addressed yet. Consequently, we evaluated the influence of DMSO on oligodendrogenesis, and on the pro-oligodendrogenic effect of MSCs’ secreted factors, using adult rat neural stem and progenitor cells (NSPCs). Here, we demonstrate that a concentration of 1% DMSO robustly suppressed oligodendrogenesis and drove the fate of differentiating NSPCs toward astrogenesis. Furthermore, the pro-oligodendrogenic effect of MSC-conditioned medium (MSCCM) was also nearly completely abolished by the presence of 1% DMSO. In this condition, inhibition of the Erk1/2 signal transduction pathway and high levels of Id2 expression, a specific inhibitor of oligodendrogenic differentiation, were detected. Furthermore, inflammatory demyelinating diseases may even potentiate the impact of DMSO on oligodendrogenesis. Our results demonstrate the imperative of considering the strong anti-oligodendrogenic activity of DMSO when designing future clinical trial protocols.
Collapse
Affiliation(s)
- Anna O'Sullivan
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria.,Department of Otorhinolaryngology, Paracelsus Medical University, Salzburg, Austria
| | - Simona Lange
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria.,Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | - Peter Rotheneichner
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria
| | - Lara Bieler
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria
| | - Ludwig Aigner
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria.,Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Francisco J Rivera
- Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria.,Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria.,Laboratory of Stem Cells and Neuroregeneration, Institute of Anatomy, Histology and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.,Center for Interdisciplinary Studies on the Nervous System (CISNe), Universidad Austral de Chile, Valdivia, Chile
| | - Sebastien Couillard-Despres
- Institute of Experimental Neuroregeneration, Paracelsus Medical University, Salzburg, Austria.,Spinal Cord Injury and Tissue Regeneration Center Salzburg (SCI-TReCS), Salzburg, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
5
|
Dunnett SB, Björklund A. Mechanisms and use of neural transplants for brain repair. PROGRESS IN BRAIN RESEARCH 2017; 230:1-51. [PMID: 28552225 DOI: 10.1016/bs.pbr.2016.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Under appropriate conditions, neural tissues transplanted into the adult mammalian brain can survive, integrate, and function so as to influence the behavior of the host, opening the prospect of repairing neuronal damage, and alleviating symptoms associated with neuronal injury or neurodegenerative disease. Alternative mechanisms of action have been postulated: nonspecific effects of surgery; neurotrophic and neuroprotective influences on disease progression and host plasticity; diffuse or locally regulated pharmacological delivery of deficient neurochemicals, neurotransmitters, or neurohormones; restitution of the neuronal and glial environment necessary for proper host neuronal support and processing; promoting local and long-distance host and graft axon growth; formation of reciprocal connections and reconstruction of local circuits within the host brain; and up to full integration and reconstruction of fully functional host neuronal networks. Analysis of neural transplants in a broad range of anatomical systems and disease models, on simple and complex classes of behavioral function and information processing, have indicated that all of these alternative mechanisms are likely to contribute in different circumstances. Thus, there is not a single or typical mode of graft function; rather grafts can and do function in multiple ways, specific to each particular context. Consequently, to develop an effective cell-based therapy, multiple dimensions must be considered: the target disease pathogenesis; the neurodegenerative basis of each type of physiological dysfunction or behavioral symptom; the nature of the repair required to alleviate or remediate the functional impairments of particular clinical relevance; and identification of a suitable cell source or delivery system, along with the site and method of implantation, that can achieve the sought for repair and recovery.
Collapse
|
6
|
Chew LJ, DeBoy CA. Pharmacological approaches to intervention in hypomyelinating and demyelinating white matter pathology. Neuropharmacology 2016; 110:605-625. [PMID: 26116759 PMCID: PMC4690794 DOI: 10.1016/j.neuropharm.2015.06.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 06/10/2015] [Accepted: 06/17/2015] [Indexed: 12/17/2022]
Abstract
White matter disease afflicts both developing and mature central nervous systems. Both cell intrinsic and extrinsic dysregulation result in profound changes in cell survival, axonal metabolism and functional performance. Experimental models of developmental white matter (WM) injury and demyelination have not only delineated mechanisms of signaling and inflammation, but have also paved the way for the discovery of pharmacological approaches to intervention. These reagents have been shown to enhance protection of the mature oligodendrocyte cell, accelerate progenitor cell recruitment and/or differentiation, or attenuate pathological stimuli arising from the inflammatory response to injury. Here we highlight reports of studies in the CNS in which compounds, namely peptides, hormones, and small molecule agonists/antagonists, have been used in experimental animal models of demyelination and neonatal brain injury that affect aspects of excitotoxicity, oligodendrocyte development and survival, and progenitor cell function, and which have been demonstrated to attenuate damage and improve WM protection in experimental models of injury. The molecular targets of these agents include growth factor and neurotransmitter receptors, morphogens and their signaling components, nuclear receptors, as well as the processes of iron transport and actin binding. By surveying the current evidence in non-immune targets of both the immature and mature WM, we aim to better understand pharmacological approaches modulating endogenous oligodendroglia that show potential for success in the contexts of developmental and adult WM pathology. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC, USA.
| | - Cynthia A DeBoy
- Biology Department, Trinity Washington University, Washington, DC, USA
| |
Collapse
|
7
|
Liu Y, Kou X, Chen C, Yu W, Su Y, Kim Y, Shi S, Liu Y. Chronic High Dose Alcohol Induces Osteopenia via Activation of mTOR Signaling in Bone Marrow Mesenchymal Stem Cells. Stem Cells 2016; 34:2157-68. [PMID: 27145264 DOI: 10.1002/stem.2392] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Revised: 03/14/2016] [Accepted: 04/14/2016] [Indexed: 12/21/2022]
Abstract
Chronic consumption of excessive alcohol results in reduced bone mass, impaired bone structure, and increased risk of bone fracture. However, the mechanisms underlying alcohol-induced osteoporosis are not fully understood. Here, we show that high dose chronic alcohol consumption reduces osteogenic differentiation and enhances adipogenic differentiation of bone marrow mesenchymal stem cells (BMMSCs), leading to osteopenia in a mouse model. Mechanistically, impaired osteo/adipogenic lineage differentiation of BMMSCs is due to activation of a phosphatidylinositide 3-kinase/AKT/mammalian target of rapamycin (mTOR) signaling cascade, resulting in downregulation of runt-related transcription factor 2 and upregulation of peroxisome proliferator-activated receptor gamma via activation of p70 ribosomal protein S6 kinase. Blockage of the mTOR pathway by rapamycin treatment ameliorates alcohol-induced osteopenia by rescuing impaired osteo/adipogenic lineage differentiation of BMMSCs. In this study, we identify a previously unknown mechanism by which alcohol impairs BMMSC lineage differentiation and reveal a potential rapamycin-based drug therapy for alcohol-induced osteoporosis. Stem Cells 2016;34:2157-2168.
Collapse
Affiliation(s)
- Yao Liu
- Department of Pediatric Dentistry, School of Stomatology, China Medical University, Shenyang, China.,Liaoning Province Key Laboratory of Oral Disease, Shenyang, China.,Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA
| | - Xiaoxing Kou
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA.,Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Chider Chen
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA.,Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Wenjing Yu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA.,Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Yingying Su
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| | - Yong Kim
- UCLA School of Dentistry, Los Angeles, California, USA
| | - Songtao Shi
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California, USA.,Department of Anatomy and Cell Biology, University of Pennsylvania, School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology, Beijing, China
| |
Collapse
|
8
|
Gesundheit B, Ashwood P, Keating A, Naor D, Melamed M, Rosenzweig JP. Therapeutic properties of mesenchymal stem cells for autism spectrum disorders. Med Hypotheses 2014; 84:169-77. [PMID: 25592283 DOI: 10.1016/j.mehy.2014.12.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 12/22/2014] [Indexed: 12/13/2022]
Abstract
Recent studies of autism spectrum disorders (ASD) highlight hyperactivity of the immune system, irregular neuronal growth and increased size and number of microglia. Though the small sample size in many of these studies limits extrapolation to all individuals with ASD, there is mounting evidence of both immune and nervous system related pathogenesis in at least a subset of patients with ASD. Given the disturbing rise in incidence rates for ASD, and the fact that no pharmacological therapy for ASD has been approved by the Food and Drug Administration (FDA), there is an urgent need for new therapeutic options. Research in the therapeutic effects of mesenchymal stem cells (MSC) for other immunological and neurological conditions has shown promising results in preclinical and even clinical studies. MSC have demonstrated the ability to suppress the immune system and to promote neurogenesis with a promising safety profile. The working hypothesis of this paper is that the potentially synergistic ability of MSC to modulate a hyperactive immune system and its ability to promote neurogenesis make it an attractive potential therapeutic option specifically for ASD. Theoretical mechanisms of action will be suggested, but further research is necessary to support these hypothetical pathways. The choice of tissue source, type of cell, and most appropriate ages for therapeutic intervention remain open questions for further consideration. Concern over poor regulatory control of stem cell studies or treatment, and the unique ethical challenges that each child with ASD presents, demands that future research be conducted with particular caution before widespread use of the proposed therapeutic intervention is implemented.
Collapse
Affiliation(s)
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California Davis, USA; Department of Medical Microbiology and Immunology, and the MIND Institute, University of California Davis, USA.
| | - Armand Keating
- Division of Hematology, University of Toronto, Cell Therapy Program, Princess Margaret Hospital, Toronto, Canada.
| | - David Naor
- Lautenberg Center for General and Tumor Immunology, Hebrew University, Hadassah Medical School, Jerusalem, Israel.
| | - Michal Melamed
- Lautenberg Center for General and Tumor Immunology, Hebrew University, Hadassah Medical School, Jerusalem, Israel.
| | | |
Collapse
|
9
|
Chen C, Akiyama K, Wang D, Xu X, Li B, Moshaverinia A, Brombacher F, Sun L, Shi S. mTOR inhibition rescues osteopenia in mice with systemic sclerosis. ACTA ACUST UNITED AC 2014; 212:73-91. [PMID: 25534817 PMCID: PMC4291526 DOI: 10.1084/jem.20140643] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Chen et al. show that treatment with rapamycin, a drug known to inhibit mTOR signaling, rescues low bone density in mice with systemic sclerosis. Fibrillin-1 (FBN1) deficiency-induced systemic sclerosis is attributed to elevation of interleukin-4 (IL4) and TGF-β, but the mechanism underlying FBN1 deficiency–associated osteopenia is not fully understood. We show that bone marrow mesenchymal stem cells (BMMSCs) from FBN1-deficient (Fbn1+/−) mice exhibit decreased osteogenic differentiation and increased adipogenic differentiation. Mechanistically, this lineage alteration is regulated by IL4/IL4Rα-mediated activation of mTOR signaling to down-regulate RUNX2 and up-regulate PPARγ2, respectively, via P70 ribosomal S6 protein kinase (P70S6K). Additionally, we reveal that activation of TGF-β/SMAD3/SP1 signaling results in enhancement of SP1 binding to the IL4Rα promoter to synergistically activate mTOR pathway in Fbn1+/− BMMSCs. Blockage of mTOR signaling by osteoblastic-specific knockout or rapamycin treatment rescues osteopenia phenotype in Fbn1+/− mice by improving osteogenic differentiation of BMMSCs. Collectively, this study identifies a previously unrecognized role of the FBN1/TGF-β/IL4Rα/mTOR cascade in BMMSC lineage selection and provides experimental evidence that rapamycin treatment may provide an anabolic therapy for osteopenia in Fbn1+/− mice.
Collapse
Affiliation(s)
- Chider Chen
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033 Department of Anatomy and Cell Biology, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Kentaro Akiyama
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033 Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Science, Kita-ku, Okayama 700-8525, Japan
| | - Dandan Wang
- Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xingtian Xu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033 Key Laboratory of Translational Research, Tong Ji University School of Stomatology, Shanghai 200072, China
| | - Bei Li
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033 School of Stomatology, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Alireza Moshaverinia
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033
| | - Frank Brombacher
- Division of Immunology, Cape Town Component and Institute of Infectious Diseases and Molecular Medicine (IIDMM), International Center for Genetic Engineering and Biotechnology (ICGEB) University of Cape Town, Cape Town 7925, South Africa
| | - Lingyun Sun
- Department of Rheumatology and Immunology, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Songtao Shi
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
10
|
Therapeutic approaches to disease modifying therapy for multiple sclerosis in adults: An Australian and New Zealand perspective Part 2 New and emerging therapies and their efficacy. J Clin Neurosci 2014; 21:1847-56. [DOI: 10.1016/j.jocn.2014.01.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/28/2014] [Indexed: 12/16/2022]
|
11
|
Chung JY, Figgett W, Fairfax K, Bernard C, Chan J, Toh BH, Mackay F, Alderuccio F. Gene therapy delivery of myelin oligodendrocyte glycoprotein (MOG) via hematopoietic stem cell transfer induces MOG-specific B cell deletion. THE JOURNAL OF IMMUNOLOGY 2014; 192:2593-601. [PMID: 24532581 DOI: 10.4049/jimmunol.1203563] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The various mechanisms that have been described for immune tolerance govern our ability to control self-reactivity and minimize autoimmunity. However, the capacity to genetically manipulate the immune system provides a powerful avenue to supplement this natural tolerance in an Ag-specific manner. We have previously shown in the mouse model of experimental autoimmune encephalomyelitis that transfer of bone marrow (BM) transduced with retrovirus encoding myelin oligodendrocyte glycoprotein (MOG) promotes disease resistance and CD4(+) T cell deletion within the thymus. However, the consequence of this strategy on B cell tolerance is not known. Using BM from IgH(MOG) mice that develop MOG-specific B cell receptors, we generated mixed chimeras together with BM-encoding MOG. In these animals, the development of MOG-specific B cells was abrogated, resulting in a lack of MOG-specific B cells in all B cell compartments examined. This finding adds a further dimension to our understanding of the mechanisms of tolerance that are associated with this gene therapy approach to treating autoimmunity and may have important implications for Ab-mediated autoimmune disorders.
Collapse
Affiliation(s)
- Jie-Yu Chung
- Department of Immunology, Central Clinical School, Monash University, Melbourne, Victoria 3181, Australia
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Cisbani G, Cicchetti F. Review: The fate of cell grafts for the treatment of Huntington's disease: thepost-mortemevidence. Neuropathol Appl Neurobiol 2014; 40:71-90. [DOI: 10.1111/nan.12104] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Accepted: 12/03/2013] [Indexed: 12/13/2022]
Affiliation(s)
- G. Cisbani
- Centre de Recherche du CHU de Québec (CHUL); Québec QC Canada
| | - F. Cicchetti
- Centre de Recherche du CHU de Québec (CHUL); Québec QC Canada
- Département de Psychiatrie et Neurosciences; Université Laval; Québec QC Canada
| |
Collapse
|
13
|
Fan C, Wang H, Chen D, Cheng X, Xiong K, Luo X, Cao Q. Effect of type-2 astrocytes on the viability of dorsal root ganglion neurons and length of neuronal processes. Neural Regen Res 2014; 9:119-128. [PMID: 25206792 PMCID: PMC4146161 DOI: 10.4103/1673-5374.125339] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2013] [Indexed: 01/06/2023] Open
Abstract
The role of type-2 astrocytes in the repair of central nervous system injury remains poorly understood. In this study, using a relatively simple culture condition in vitro, type-2 astrocytes, differentiated from oligodendrocyte precursor cells by induction with bone morphogenetic protein-4, were co-cultured with dorsal root ganglion neurons. We examined the effects of type-2 astrocytes differentiated from oligodendrocyte precursor cells on the survival and growth of dorsal root ganglion neurons. Results demonstrated that the number of dorsal root ganglion neurons was higher following co-culture of oligodendrocyte precursor cells and type-2 astrocytes than when cultured alone, but lower than that of neurons co-cultured with type-1 astrocytes. The length of the longest process and the length of all processes of a single neuron were shortest in neurons cultured alone, followed by neurons co-cultured with type-2 astrocytes, then neurons co-cultured with oligodendrocyte precursor cells, and longest in neurons co-cultured with type-1 astrocytes. These results indicate that co-culture with type-2 astrocytes can increase neuronal survival rate and process length. However, compared with type-1 astrocytes and oligodendrocyte precursor cells, the promotion effects of type-2 astrocytes on the growth of dorsal root ganglion neurons were weaker.
Collapse
Affiliation(s)
- Chunling Fan
- Department of Human Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Hui Wang
- Department of Human Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Dan Chen
- Department of Human Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Xiaoxin Cheng
- The Vivian L Smith Department of Neurosurgery, UT Medical School at Houston, Houston, TX, USA
| | - Kun Xiong
- Department of Human Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Xuegang Luo
- Department of Human Anatomy and Neurobiology, Xiangya School of Medicine, Central South University, Changsha, Hunan Province, China
| | - Qilin Cao
- The Vivian L Smith Department of Neurosurgery, UT Medical School at Houston, Houston, TX, USA
| |
Collapse
|
14
|
Intrathecal Mesenchymal Stem Cell Therapy in Multiple Sclerosis: A Follow-Up Study for Five Years After Injection. ARCHIVES OF NEUROSCIENCE 2013. [DOI: 10.5812/archneurosci.13687] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
15
|
Derivation of Pre-oligodendrocytes from Human Endometrial Stromal Cells by Using Overexpression of MicroRNA 338. J Mol Neurosci 2013; 51:337-43. [DOI: 10.1007/s12031-013-0101-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 08/15/2013] [Indexed: 11/29/2022]
|
16
|
Programming of human endometrial-derived stromal cells (EnSCs) into pre-oligodendrocyte cells by overexpression of miR-219. Neurosci Lett 2013; 537:65-70. [DOI: 10.1016/j.neulet.2013.01.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/15/2013] [Accepted: 01/17/2013] [Indexed: 01/01/2023]
|
17
|
Giunti D, Parodi B, Usai C, Vergani L, Casazza S, Bruzzone S, Mancardi G, Uccelli A. Mesenchymal stem cells shape microglia effector functions through the release of CX3CL1. Stem Cells 2013; 30:2044-53. [PMID: 22821677 DOI: 10.1002/stem.1174] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSC) display a remarkable ability to modulate the immune response and protect the central nervous system mainly through the release of soluble factors in a paracrine fashion, affecting the functional behavior of cells in the tissues. Here we investigated the effect of the interaction between MSC and microglia in vitro, and we dissected the molecular and cellular mechanisms of this crosstalk. We demonstrated that MSC impair microglia activation by inflammatory cues through the inhibition of the expression and release of inflammatory molecules and stress-associated proteins. We showed that MSC significantly increase microglial expression and release of molecules associated with a neuroprotective phenotype such as CX3CR1, nuclear receptor 4 family, CD200 receptor, and insulin growth factor 1. Interestingly, MSC can enhance functional changes on microglia as depicted by the increase of intracellular calcium concentration and phagocytic activity. This last event is associated with an increased expression of triggering receptor expressed on myeloid cells-2, an innate immune receptor involved in phagocytosis in the absence of inflammation. The observed effects on CX3CR1-expressing microglia are due to the release of CX3CL1 by MSC, driven by inflammatory signals, as demonstrated by the reversal of the observed results when CX3CL1 expression was silenced in MSC or its release was blocked. Finally, we showed that exogenous CX3CL1 induce phenotypic and functional changes of microglia similar to those induced by MSC. These findings demonstrate that MSC instruct, through the release of CX3CL1, microglia responsiveness to proinflammatory signals by modulating constitutive "calming" receptors, typically expressed by "steady-state microglia" thus switching microglia from a detrimental phenotype to a neuroprotective one.
Collapse
Affiliation(s)
- Debora Giunti
- Department of Neurosciences, Ophthalmology and Genetics, University of Genoa, Genoa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Ebrahimi-Barough S, Kouchesfahani HM, Ai J, Massumi M. Differentiation of human endometrial stromal cells into oligodendrocyte progenitor cells (OPCs). J Mol Neurosci 2013; 51:265-73. [PMID: 23338937 DOI: 10.1007/s12031-013-9957-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 01/09/2013] [Indexed: 01/21/2023]
Abstract
Oligodendrocytes are myelinating cells in the central nervous system that form the myelin sheath of axons to support rapid nerve conduction. Human endometrial stromal cells (EnSCs) are the abundant and easy available source for cell replacement therapy. In the present study, the EnSCs were coaxed to oligodendrocyte progenitor programming by induction of neuronal condition media, including bFGF, epidermal growth factor, and platelet-derived growth factor (PDGF)-AA signaling molecules as well as triiodothyronine. Differentiated cells were analyzed for expression of oligodendrocytic markers by quantitative reverse transcription PCR and immunocytochemistry. The results showed the expression of oligodendrocyte lineage markers such as nestin, PDGF receptor alpha (PDGFRα), Sox10, and Olig2 in the level of mRNAs. The expression of nestin and PDGFRα increased after 8 days posttreatment. Interestingly, the expression of nestin and PDGFRα genes at the levels of mRNA and proteins decreased 24 days after induction. The expression of A2B5, O4, and Olig2 proteins in EnSCs was confirmed using immunocytochemistry. The results confirmed that EnSCs could response to the signaling molecules which routinely applied for oligodendrocyte differentiation. Here for the first time, we demonstrated that EnSCs could be programmed into oligodendrocyte progenitor cells and may convince to consider these cells as suitable source for cell therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Somayeh Ebrahimi-Barough
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University (TMU), Tehran, Iran,
| | | | | | | |
Collapse
|
19
|
Rutten MJ, Janes MA, Chang IR, Gregory CR, Gregory KW. Development of a functional schwann cell phenotype from autologous porcine bone marrow mononuclear cells for nerve repair. Stem Cells Int 2012; 2012:738484. [PMID: 22792117 PMCID: PMC3388598 DOI: 10.1155/2012/738484] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Accepted: 03/29/2012] [Indexed: 01/10/2023] Open
Abstract
Adult bone marrow mononuclear cells (BM-MNCs) are a potential resource for making Schwann cells to repair damaged peripheral nerves. However, many methods of producing Schwann-like cells can be laborious with the cells lacking a functional phenotype. The objective of this study was to develop a simple and rapid method using autologous BM-MNCs to produce a phenotypic and functional Schwann-like cell. Adult porcine bone marrow was collected and enriched for BM-MNCs using a SEPAX device, then cells cultured in Neurobasal media, 4 mM L-glutamine and 20% serum. After 6-8 days, the cultures expressed Schwann cell markers, S-100, O4, GFAP, were FluoroMyelin positive, but had low p75(NGF) expression. Addition of neuregulin (1-25 nM) increased p75(NGF) levels at 24-48 hrs. We found ATP dose-dependently increased intracellular calcium [Ca(2+)](i), with nucleotide potency being UTP = ATP > ADP > AMP > adenosine. Suramin blocked the ATP-induced [Ca(2+)](i) but α, β,-methylene-ATP had little effect suggesting an ATP purinergic P2Y2 G-protein-coupled receptor is present. Both the Schwann cell markers and ATP-induced [Ca(2+)](i) sensitivity decreased in cells passaged >20 times. Our studies indicate that autologous BM-MNCs can be induced to form a phenotypic and functional Schwann-like cell which could be used for peripheral nerve repair.
Collapse
Affiliation(s)
- Michael J. Rutten
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
- OHSU Center for Regenerative Medicine, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
| | - Michael Ann Janes
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
| | - Ivy R. Chang
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
| | - Cynthia R. Gregory
- OHSU Center for Regenerative Medicine, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
- Oregon Biomedical Engineering Institute, 25999 SW Canyon Creek Rd., Wilsonville, OR 97070, USA
- Portland VA Medical Center, 3710 SW U.S. Veterans Hospital Rd., Portland, OR 97239, USA
| | - Kenton W. Gregory
- Providence Health and Services, 9555 SW Barnes Rd., Portland, OR 97225, USA
- OHSU Center for Regenerative Medicine, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Road, Portland, OR 97239, USA
- Oregon Biomedical Engineering Institute, 25999 SW Canyon Creek Rd., Wilsonville, OR 97070, USA
| |
Collapse
|
20
|
Garbossa D, Boido M, Fontanella M, Fronda C, Ducati A, Vercelli A. Recent therapeutic strategies for spinal cord injury treatment: possible role of stem cells. Neurosurg Rev 2012; 35:293-311; discussion 311. [PMID: 22539011 DOI: 10.1007/s10143-012-0385-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 09/27/2011] [Accepted: 11/20/2011] [Indexed: 01/01/2023]
Abstract
Spinal cord injury (SCI) often results in significant dysfunction and disability. A series of treatments have been proposed to prevent and overcome the formation of the glial scar and inhibitory factors to axon regrowth. In the last decade, cell therapy has emerged as a new tool for several diseases of the nervous system. Stem cells act as minipumps providing trophic and immunomodulatory factors to enhance axonal growth, to modulate the environment, and to reduce neuroinflammation. This capability can be boosted by genetical manipulation to deliver trophic molecules. Different types of stem cells have been tested, according to their properties and the therapeutic aims. They differ from each other for origin, developmental stage, stage of differentiation, and fate lineage. Related to this, stem cells differentiating into neurons could be used for cell replacement, even though the feasibility that stem cells after transplantation in the adult lesioned spinal cord can differentiate into neurons, integrate within neural circuits, and emit axons reaching the muscle is quite remote. The timing of cell therapy has been variable, and may be summarized in the acute and chronic phases of disease, when stem cells interact with a completely different environment. Even though further experimental studies are needed to elucidate the mechanisms of action, the therapeutic, and the side effects of cell therapy, several clinical protocols have been tested or are under trial. Here, we report the state-of-the-art of cell therapy in SCI, in terms of feasibility, outcome, and side effects.
Collapse
Affiliation(s)
- D Garbossa
- Department of Neurosurgery, S. Giovanni Battista Hospital, University of Torino, Via Cherasco 15, 10126, Torino, Italy.
| | | | | | | | | | | |
Collapse
|
21
|
Parenteau N, Hardin-Young J, Shannon W, Cantini P, Russell A. Meeting the need for regenerative therapies I: target-based incidence and its relationship to U.S. spending, productivity, and innovation. TISSUE ENGINEERING. PART B, REVIEWS 2012; 18:139-54. [PMID: 22044424 PMCID: PMC3311404 DOI: 10.1089/ten.teb.2011.0454] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Accepted: 11/01/2011] [Indexed: 12/13/2022]
Abstract
Regenerative therapies possess high theoretical potential for medical advance yet their success as commercial therapeutics is still open to debate. Appropriate data on target opportunities that provide perspective and enable strategic decision making is necessary for both efficient and effective translation. Up until now, this data have been out of reach to research scientists and many start-up companies-the very groups currently looked to for the critical advance of these therapies. The target-based estimate of opportunity presented in this report demonstrates its importance in evaluating medical need and technology feasibility. In addition, analysis of U.S. research spending, productivity, and innovation reveals that U.S. basic research in this field would benefit from greater interdisciplinarity. Overcoming the barriers that currently prevent translation into high value therapies that are quickly clinically adopted requires simultaneous integration of engineering, science, business, and clinical practice. Achieving this integration is nontrivial.
Collapse
Affiliation(s)
| | | | - William Shannon
- BioRankings, LLC, St. Louis, Missouri
- Division of General Medical Sciences, Washington University School of Medicine, St. Louis, Missouri
| | - Patrick Cantini
- The McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania
| | - Alan Russell
- The McGowan Institute of Regenerative Medicine, Pittsburgh, Pennsylvania
- Department of Surgery, University of Pittsburgh Medical Center, UPMC Presbyterian, Pittsburgh, Pennsylvania
| |
Collapse
|
22
|
Electric impedance sensing in cell-substrates for rapid and selective multipotential differentiation capacity monitoring of human mesenchymal stem cells. Biosens Bioelectron 2012; 34:63-9. [DOI: 10.1016/j.bios.2012.01.013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Revised: 01/11/2012] [Accepted: 01/13/2012] [Indexed: 01/18/2023]
|
23
|
Mauri M, Lentini D, Gravati M, Foudah D, Biella G, Costa B, Toselli M, Parenti M, Coco S. Mesenchymal stem cells enhance GABAergic transmission in co-cultured hippocampal neurons. Mol Cell Neurosci 2012; 49:395-405. [PMID: 22388097 DOI: 10.1016/j.mcn.2012.02.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 02/14/2012] [Accepted: 02/15/2012] [Indexed: 12/14/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) are multipotent stem cells endowed with neurotrophic potential combined with immunological properties, making them a promising therapeutic tool for neurodegenerative disorders. However, the mechanisms through which MSCs promote the neurological recovery following injury or inflammation are still largely unknown, although cell replacement and paracrine mechanisms have been hypothesized. In order to find out what are the mechanisms of the trophic action of MSCs, as compared to glial cells, on CNS neurons, we set up a co-culture system where rat MSCs (or cortical astrocytes) were used as a feeding layer for hippocampal neurons without any direct contact between the two cell types. The analysis of hippocampal synaptogenesis, synaptic vesicle recycling and electrical activity show that MSCs were capable to support morphological and functional neuronal differentiation. The proliferation of hippocampal glial cells induced by the release of bioactive substance(s) from MSCs was necessary for neuronal survival. Furthermore, MSCs selectively increased hippocampal GABAergic pre-synapses. This effect was paralleled with a higher expression of the potassium/chloride KCC2 co-transporter and increased frequency and amplitude of mIPSCs and sIPSCs. The enhancement of GABA synapses was impaired by the treatment with K252a, a Trk/neurotrophin receptor blocker, and by TrkB receptor bodies hence suggesting the involvement of BDNF as a mediator of such effects. The results obtained here indicate that MSC-secreted factors induce glial-dependent neuronal survival and trigger an augmented GABAergic transmission in hippocampal cultures, highlighting a new effect by which MSCs could promote CNS repair. Our results suggest that MSCs may be useful in those neurological disorders characterized by an impairment of excitation versus inhibition balance.
Collapse
Affiliation(s)
- Mario Mauri
- Department of Experimental Medicine, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
The use of pluripotent stem cell for personalized cell therapies against neurological disorders. J Biomed Biotechnol 2011; 2011:520816. [PMID: 22203784 PMCID: PMC3238807 DOI: 10.1155/2011/520816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Accepted: 11/04/2011] [Indexed: 11/18/2022] Open
Abstract
Although there are a number of weaknesses for clinical use, pluripotent stem cells are valuable sources for patient-specific cell therapies against various diseases. Backed-up by a huge number of basic researches, neuronal differentiation mechanism is well established and pluripotent stem cell therapies against neurological disorders are getting closer to clinical application. However, there are increasing needs for standardization of the sourcing pluripotent stem cells by establishing stem cell registries and banking. Global harmonization will accelerate practical use of personalized therapies using pluripotent stem cells.
Collapse
|
25
|
Natalizumab in aggressive multiple sclerosis after haematopoietic stem cell transplantation. Neurol Sci 2011; 33:863-7. [DOI: 10.1007/s10072-011-0848-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2010] [Accepted: 10/28/2011] [Indexed: 10/15/2022]
|
26
|
Durnaoglu S, Genc S, Genc K. Patient-specific pluripotent stem cells in neurological diseases. Stem Cells Int 2011; 2011:212487. [PMID: 21776279 PMCID: PMC3138107 DOI: 10.4061/2011/212487] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2010] [Revised: 03/28/2011] [Accepted: 05/09/2011] [Indexed: 12/29/2022] Open
Abstract
Many human neurological diseases are not currently curable and result in devastating neurologic sequelae. The increasing availability of induced pluripotent stem cells (iPSCs) derived from adult human somatic cells provides new prospects for cellreplacement strategies and disease-related basic research in a broad spectrum of human neurologic diseases. Patient-specific iPSC-based modeling of neurogenetic and neurodegenerative diseases is an emerging efficient tool for in vitro modeling to understand disease and to screen for genes and drugs that modify the disease process. With the exponential increase in iPSC research in recent years, human iPSCs have been successfully derived with different technologies and from various cell types. Although there remain a great deal to learn about patient-specific iPSC safety, the reprogramming mechanisms, better ways to direct a specific reprogramming, ideal cell source for cellular grafts, and the mechanisms by which transplanted stem cells lead to an enhanced functional recovery and structural reorganization, the discovery of the therapeutic potential of iPSCs offers new opportunities for the treatment of incurable neurologic diseases. However, iPSC-based therapeutic strategies need to be thoroughly evaluated in preclinical animal models of neurological diseases before they can be applied in a clinical setting.
Collapse
Affiliation(s)
- Serpen Durnaoglu
- Department of Neuroscience, Health Science Institute, Dokuz Eylül University, Inciralti, 35340 Izmir, Turkey
| | | | | |
Collapse
|
27
|
Current World Literature. Curr Opin Neurol 2011; 24:300-7. [DOI: 10.1097/wco.0b013e328347b40e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
28
|
Zeher M, Papp G, Szodoray P. Autologous haemopoietic stem cell transplantation for autoimmune diseases. Expert Opin Biol Ther 2011; 11:1193-201. [PMID: 21609185 DOI: 10.1517/14712598.2011.580272] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
29
|
Forraz N, McGuckin CP. The umbilical cord: a rich and ethical stem cell source to advance regenerative medicine. Cell Prolif 2011; 44 Suppl 1:60-9. [PMID: 21481046 PMCID: PMC6495455 DOI: 10.1111/j.1365-2184.2010.00729.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Accepted: 08/11/2010] [Indexed: 02/04/2023] Open
Abstract
Science and medicine place a lot of hope in the development of stem cell research and regenerative medicine. This review will define the concept of regenerative medicine and focus on an abundant stem cell source - neonatal tissues such as the umbilical cord. Umbilical cord blood has been used clinically for over 20 years as a cell source for haematopoietic stem cell transplantation. Beyond this, cord blood and umbilical cord-derived stem cells have demonstrated potential for pluripotent lineage differentiation (liver, pancreatic, neural tissues and more) in vitro and in vivo. This promising research has opened up a new era for utilization of neonatal stem cells, now used beyond haematology in clinical trials for autoimmune disorders, cerebral palsy or type I diabetes.
Collapse
Affiliation(s)
- N. Forraz
- CTI‐LYON, Cell Therapy Research Institute, Parc Technologique de Lyon St Priest, St Priest‐Lyon, France
| | - C. P. McGuckin
- CTI‐LYON, Cell Therapy Research Institute, Parc Technologique de Lyon St Priest, St Priest‐Lyon, France
| |
Collapse
|
30
|
Current world literature. Curr Opin Oncol 2011; 23:227-34. [PMID: 21307677 DOI: 10.1097/cco.0b013e328344b687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
31
|
Siatskas C, Payne NL, Short MA, Bernard CCA. A consensus statement addressing mesenchymal stem cell transplantation for multiple sclerosis: it's time! Stem Cell Rev Rep 2011; 6:500-6. [PMID: 20665128 DOI: 10.1007/s12015-010-9173-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple sclerosis is a neurodegenerative disease of the central nervous system that is characterized by inflammation, demyelination with associated accumulation of myelin debris, oligodendrocyte and axonal loss. Current therapeutic interventions for multiple sclerosis predominantly modulate the immune system and reduce the inflammatory insult by general, non-specific mechanisms but have little effect on the neurodegenerative component of the disease. Predictably, the overall long-term impact of treatment is limited since the neurodegenerative component of the disease, which can be the dominant process in some patients, determines permanent disability. Mesenchymal stem cells, which are endowed with potent immune regulatory and neuroprotective properties, have recently emerged as promising cellular vehicles for the treatment of MS. Preclinical evaluation in experimental models of MS have shown that MSCs are efficacious in suppressing clinical disease. Mechanisms that may underlie these effects predominantly involve the secretion of immunomodulatory and neurotrophic growth factors, which collectively act to limit CNS inflammation, stimulate neurogenesis, protect axons and promote remyelination. As a logical progression to clinical utility, the safety of these cells have been initially assessed in hematological, cardiac and inflammatory diseases. Importantly, transplantation with autologous or allogeneic MSCs has been well tolerated by patients with few significant adverse effects. On the basis of these results, new, multicentre clinical trials have been launched to assess the safety and efficacy of MSCs for inflammatory MS. It thus comes as no surprise that the coalescence of an international group of experts have convened to generate a consensus guide for the transplantation of autologous bone marrow-derived MSC which, in time, may set the foundation for the next generation of therapies for the treatment of MS patients.
Collapse
Affiliation(s)
- Christopher Siatskas
- Monash immunology and stem cell laboratories, Monash University, Clayton, Victoria, 3800, Australia.
| | | | | | | |
Collapse
|
32
|
Dunnett SB, Rosser AE. Cell-based treatments for huntington's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2011; 98:483-508. [PMID: 21907097 DOI: 10.1016/b978-0-12-381328-2.00017-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In experimental rats, mice, and monkeys, transplantation of embryonic striatal cells into the striatum can repair the damage and alleviate the functional deficits caused by striatal lesions. Such strategies have been translated to striatal repair by cell transplantation in small numbers of patients with progressive genetic striatal degeneration in Huntington's disease. In spite of some encouraging preliminary data, the clinical results are to date neither as reliable nor as compelling as the broad extend of recovery observed in the animal models across motor, cognitive, and skill and habit learning domains. Strategies to achieve immediate and long-term improvements in the clinical applications include identifying and limiting the causes of complications, standardization and quality control of preparation and delivery, appropriate patient selection to match the cellular repair to specific profiles of cell loss and degeneration in individual patients and different neurodegenerative diseases, and improving the availability of alternative sources of donor cells and tissues.
Collapse
Affiliation(s)
- Stephen B Dunnett
- Brain Repair Group, Schools of Biosciences and Medicine, Cardiff University, Cardiff, Wales, UK
| | | |
Collapse
|
33
|
Bone marrow mesenchymal stem cell transplantation in patients with multiple sclerosis: a pilot study. J Neuroimmunol 2010; 227:185-9. [PMID: 20728948 DOI: 10.1016/j.jneuroim.2010.07.013] [Citation(s) in RCA: 261] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 07/12/2010] [Accepted: 07/19/2010] [Indexed: 12/13/2022]
Abstract
We explore the safety, and therapeutic benefit of intrathecal injection of ex-vivo expanded autologous bone marrow derived mesenchymal stem cells (BM-MSCs) in 10 patients with advanced multiple sclerosis (MS). Patients were assessed at 3, 6 and 12 months. Assessment at 3-6 months revealed Expanded Disability Scale Score (EDSS) improvement in 5/7, stabilization in 1/7, and worsening in 1/7 patients. MRI at 3 months revealed new or enlarging lesions in 5/7 and Gadolinium (Gd+) enhancing lesions in 3/7 patients. Vision and low contrast sensitivity testing at 3 months showed improvement in 5/6 and worsening in 1/6 patients. Early results show hints of clinical but not radiological efficacy and evidence of safety with no serious adverse events.
Collapse
|
34
|
Tokumoto Y, Ogawa S, Nagamune T, Miyake J. Comparison of efficiency of terminal differentiation of oligodendrocytes from induced pluripotent stem cells versus embryonic stem cells in vitro. J Biosci Bioeng 2009; 59:882-92. [PMID: 20471604 DOI: 10.1002/glia.21159] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 01/19/2011] [Indexed: 12/28/2022]
Abstract
Oligodendrocytes are the myelinating cells of the central nervous system (CNS), and defects in these cells can result in the loss of CNS functions. Although oligodendrocyte progenitor cells transplantation therapy is an effective cure for such symptoms, there is no readily available source of these cells. Recent studies have described the generation of induced pluripotent stem cells (iPS cells) from somatic cells, leading to anticipation of this technique as a novel therapeutic tool in regenerative medicine. In this study, we evaluated the ability of iPS cells derived from mouse embryonic fibroblasts to differentiate into oligodendrocytes and compared this with the differential ability of mouse embryonic stem cells (ES cells). Experiments using an in vitro oligodendrocyte differentiation protocol that was optimized to ES cells demonstrated that 2.3% of iPS cells differentiated into O4(+) oligodendrocytes compared with 24.0% of ES cells. However, the rate of induction of A2B5(+) oligodendrocyte precursor cell (OPC) was similar for both iPS-derived cells and ES-derived cells (14.1% and 12.6%, respectively). These findings suggest that some intracellular factors in iPS cells inhibit the terminal differentiation of oligodendrocytes from the OPC stage.
Collapse
Affiliation(s)
- Yasuhito Tokumoto
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | | | | | | |
Collapse
|