1
|
Gavasso S, Kråkenes T, Olsen H, Evjenth EC, Ytterdal M, Haugsøen JB, Kvistad CE. The Therapeutic Mechanisms of Mesenchymal Stem Cells in MS-A Review Focusing on Neuroprotective Properties. Int J Mol Sci 2024; 25:1365. [PMID: 38338644 PMCID: PMC10855165 DOI: 10.3390/ijms25031365] [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: 12/08/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
In multiple sclerosis (MS), there is a great need for treatment with the ability to suppress compartmentalized inflammation within the central nervous system (CNS) and to promote remyelination and regeneration. Mesenchymal stem cells (MSCs) represent a promising therapeutic option, as they have been shown to migrate to the site of CNS injury and exert neuroprotective properties, including immunomodulation, neurotrophic factor secretion, and endogenous neural stem cell stimulation. This review summarizes the current understanding of the underlying neuroprotective mechanisms and discusses the translation of MSC transplantation and their derivatives from pre-clinical demyelinating models to clinical trials with MS patients.
Collapse
Affiliation(s)
- Sonia Gavasso
- Department of Clinical Medicine, University of Bergen, 5009 Bergen, Norway; (T.K.); (H.O.); (E.C.E.); (J.B.H.); (C.E.K.)
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Torbjørn Kråkenes
- Department of Clinical Medicine, University of Bergen, 5009 Bergen, Norway; (T.K.); (H.O.); (E.C.E.); (J.B.H.); (C.E.K.)
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Håkon Olsen
- Department of Clinical Medicine, University of Bergen, 5009 Bergen, Norway; (T.K.); (H.O.); (E.C.E.); (J.B.H.); (C.E.K.)
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Elisabeth Claire Evjenth
- Department of Clinical Medicine, University of Bergen, 5009 Bergen, Norway; (T.K.); (H.O.); (E.C.E.); (J.B.H.); (C.E.K.)
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Marie Ytterdal
- Department of Clinical Medicine, University of Bergen, 5009 Bergen, Norway; (T.K.); (H.O.); (E.C.E.); (J.B.H.); (C.E.K.)
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Jonas Bull Haugsøen
- Department of Clinical Medicine, University of Bergen, 5009 Bergen, Norway; (T.K.); (H.O.); (E.C.E.); (J.B.H.); (C.E.K.)
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| | - Christopher Elnan Kvistad
- Department of Clinical Medicine, University of Bergen, 5009 Bergen, Norway; (T.K.); (H.O.); (E.C.E.); (J.B.H.); (C.E.K.)
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway
| |
Collapse
|
2
|
Ehsani A, Jodaei A, Barzegar-Jalali M, Fathi E, Farahzadi R, Adibkia K. Nanomaterials and Stem Cell Differentiation Potential: An Overview of Biological Aspects and Biomedical Efficacy. Curr Med Chem 2021; 29:1804-1823. [PMID: 34254903 DOI: 10.2174/0929867328666210712193113] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 11/22/2022]
Abstract
Nanoparticles (NPs) due to their medical applications are widely used. Accordingly, the use of mesenchymal stem cells is one of the most important alternatives in tissue engineering field. NPs play effective roles in stem cells proliferation and differentiation. The combination of NPs and tissue regeneration by stem cells has created new therapeutic approach towards humanity. Of note, the physicochemical properties of NPs determine their biological function. Interestingly, various mechanisms such as modulation of signaling pathways and generation of reactive oxygen species, are involved in NPs-induced cellular proliferation and differentiation. This review summarized the types of nanomaterials effective on stem cell differentiation, the physicochemical features, biomedical application of these materials and relationship between nanomaterials and environment.
Collapse
Affiliation(s)
- Ali Ehsani
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asma Jodaei
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Ezzatollah Fathi
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khosro Adibkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Wang F, Zhu J, Zheng J, Duan W, Zhou Z. miR‑210 enhances mesenchymal stem cell‑modulated neural precursor cell migration. Mol Med Rep 2020; 21:2405-2414. [PMID: 32323777 PMCID: PMC7185297 DOI: 10.3892/mmr.2020.11065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 03/15/2018] [Indexed: 11/06/2022] Open
Abstract
The migration of endogenous neural stem cells and neural precursor cells (NPCs) to sites of injury is essential for neuroregeneration following hypoxic‑ischemic events. Bone marrow‑derived mesenchymal stem cells (BMSCs) are a potential therapeutic source of cells following central nervous system damage; however, few studies have investigated the effects of BMSCs on cell migration. Thus, in the present study, the effects of BMSCs on NPC migration were investigated. In the present study, BMSCs and NPCs were isolated and cultured from mice. The effects of BMSCs on the migration of NPCs were analyzed using a Transwell cell migration assay. BMSCs were transfected with microRNA‑210 (miR‑210) mimics and inhibitors to examine the effects of the respective upregulation and downregulation of miR‑210 in BMSCs on the migration of NPCs. Then, miR‑210 expression in BMSCs were quantified and the expression levels of vascular endothelial growth factor‑C (VEGF‑C), brain derived neurotrophic factor (BDNF) and chemokine C‑C motif ligand 3 (CCL3) in the supernatant under hypoxic conditions were investigated via reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and ELISA. Subsequently, the expression of VEGF‑C, BDNF and CCL3 in BMSCs overexpressing miR‑210 or BMSCs suppressing miR‑210 was examined by RT‑qPCR and western blot analyses. BMSCs promoted the migration of NPC, particularly when pre‑cultured with BMSCs for 24 h and co‑cultured with NPCs for 24 h; the miR‑210 expression levels increased under hypoxic conditions. Additionally, the migration of NPCs was also increased when the BMSCs overexpressed miR‑210 compared with the BMSCs transfected with a negative control miR and BMSCs with downregulated miR‑210 levels. The expression levels of VEGF‑C increased in the BMSCs that overexpressed miR‑210 and were decreased in BMSCs transfected with a miR‑210 inhibitor. The results of the present study indicated that BMSCs promote the migration of NPCs. Overexpression of miR‑210 in BMSCs enhanced NPC migration and may be associated with increases in VEGF‑C expression levels.
Collapse
Affiliation(s)
- Faxiang Wang
- Department of Neurology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, P.R. China
| | - Jie Zhu
- Department of Neurology, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing 400042, P.R. China
| | - Jian Zheng
- Department of Neurology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, P.R. China
| | - Wei Duan
- Department of Neurology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, P.R. China
| | - Zhujuan Zhou
- Department of Neurology, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, P.R. China
| |
Collapse
|
4
|
Katheria AC, Amino R, Konop JM, Orona AJ, Kim E, Liu Y, Wu D, Snyder EY. Stem Cell Composition of Umbilical Cord Blood Following Milking Compared with Delayed Clamping of the Cord Appears Better Suited for Promoting Hematopoiesis. J Pediatr 2020; 216:222-226. [PMID: 31471115 DOI: 10.1016/j.jpeds.2019.07.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/06/2019] [Accepted: 07/15/2019] [Indexed: 12/23/2022]
Abstract
In comparing placental transfusion strategies, blood obtained from an umbilical cord that has been "milked" vs one in which clamping was simply delayed contains mesenchymal stromal cells in addition to solely hematopoietic stem cells, a composition more favorable for hematopoiesis, as suggested by its superior rescue of lethally irradiated bone marrow-depleted mice.
Collapse
Affiliation(s)
- Anup C Katheria
- Neonatal Research Institute, Sharp Mary Birch Hospital for Women and Newborns, San Diego, CA.
| | - Ramina Amino
- Department of Pediatrics, University of California San Diego, La Jolla, CA; Center for Stem Cells and Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA; Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Jason M Konop
- Center for Stem Cells and Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA; Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Anthony J Orona
- Center for Stem Cells and Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA; Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Elizabeth Kim
- Center for Stem Cells and Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Yang Liu
- Center for Stem Cells and Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA; Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Donlaong Wu
- Center for Stem Cells and Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA; Sanford Consortium for Regenerative Medicine, La Jolla, CA
| | - Evan Y Snyder
- Department of Pediatrics, University of California San Diego, La Jolla, CA; Center for Stem Cells and Regenerative Medicine, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA; Sanford Consortium for Regenerative Medicine, La Jolla, CA.
| |
Collapse
|
5
|
Wilson JJ, Foyle KL, Foeng J, Norton T, McKenzie DR, Payne N, Bernard CC, McColl SR, Comerford I. Redirecting adult mesenchymal stromal cells to the brain: a new approach for treating CNS autoimmunity and neuroinflammation? Immunol Cell Biol 2018; 96:347-357. [PMID: 29377354 DOI: 10.1111/imcb.12014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 12/15/2022]
Abstract
Mesenchymal stromal cells or stem cells (MSCs) have been shown to participate in tissue repair and are immunomodulatory in neuropathological settings. Given this, their potential use in developing a new generation of personalized therapies for autoimmune and inflammatory diseases of the central nervous system (CNS) will be explored. To effectively exert these effector functions, MSCs must first gain entry into damaged neural tissues, a process that has been demonstrated to be a limiting factor in their therapeutic efficacy. In this review, we discuss approaches to maximize the therapeutic efficacy of MSCs by altering their intrinsic trafficking programs to effectively enter neuropathological sites. To this end, we explore the significant role of chemokine receptors and adhesion molecules in directing cellular traffic to the inflamed CNS and the capacity of MSCs to adopt these molecular mechanisms to gain entry to this site. We postulate that understanding and exploiting these migratory mechanisms may be key to the development of cell-based therapies tailored to respond to the migratory cues unique to the nature and stage of progression of individual CNS disorders.
Collapse
Affiliation(s)
- Jasmine J Wilson
- The Chemokine Biology Laboratory, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Kerrie L Foyle
- The Chemokine Biology Laboratory, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jade Foeng
- The Chemokine Biology Laboratory, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Todd Norton
- The Chemokine Biology Laboratory, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Duncan R McKenzie
- The Chemokine Biology Laboratory, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Natalie Payne
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Claude C Bernard
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Shaun R McColl
- The Chemokine Biology Laboratory, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Iain Comerford
- The Chemokine Biology Laboratory, The University of Adelaide, Adelaide, SA, 5005, Australia
| |
Collapse
|
6
|
Köse S, Kankilic B, Gizer M, Ciftci Dede E, Bayramli E, Korkusuz P, Korkusuz F. Stem Cell and Advanced Nano Bioceramic Interactions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1077:317-342. [PMID: 30357696 DOI: 10.1007/978-981-13-0947-2_17] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Bioceramics are type of biomaterials generally used for orthopaedic applications due to their similar structure with bone. Especially regarding to their osteoinductivity and osteoconductivity, they are used as biodegradable scaffolds for bone regeneration along with mesenchymal stem cells. Since chemical properties of bioceramics are important for regeneration of tissue, physical properties are also important for cell proliferation. In this respect, several different manufacturing methods are used for manufacturing nano scale bioceramics. These nano scale bioceramics are used for regeneration of bone and cartilage both alone or with other types of biomaterials. They can also act as carrier for the delivery of drugs in musculoskeletal infections without causing any systemic toxicity.
Collapse
Affiliation(s)
- Sevil Köse
- Faculty of Health Sciences, Department of Nutrition and Dietetics, Atilim University, Ankara, Turkey.
| | - Berna Kankilic
- Head of Certification, Directorate of Directives, Turkish Standards Institution, Ankara, Turkey
| | - Merve Gizer
- Department of Bioengineering, Hacettepe University, Ankara, Turkey
| | - Eda Ciftci Dede
- Department of Bioengineering, Hacettepe University, Ankara, Turkey
| | - Erdal Bayramli
- Department of Chemistry, Middle East Technical University, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Feza Korkusuz
- Department of Sports Medicine, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
7
|
Three-Dimensional Organoid System Transplantation Technologies in Future Treatment of Central Nervous System Diseases. Stem Cells Int 2017; 2017:5682354. [PMID: 28904534 PMCID: PMC5585580 DOI: 10.1155/2017/5682354] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/24/2017] [Accepted: 06/08/2017] [Indexed: 02/06/2023] Open
Abstract
In recent years, scientists have made great achievements in understanding the development of human brain and elucidating critical elements of stepwise spatiotemporal control strategies in neural stem cell specification lineage, which facilitates successful induction of neural organoid in vitro including the cerebral cortex, cerebellar, neural tube, hippocampus cortex, pituitary, and optic cup. Besides, emerging researches on neural organogenesis promote the application of 3D organoid system transplantation in treating central nervous system (CNS) diseases. Present review will categorize current researches on organogenesis into three approaches: (a) stepwise, direct organization of region-specific or population-enriched neural organoid; (b) assemble and direct distinct organ-specific progenitor cells or stem cells to form specific morphogenesis organoid; and (c) assemble embryoid bodies for induction of multilayer organoid. However, the majority of these researches focus on elucidating cellular and molecular mechanisms involving in brain organogenesis or disease development and only a few of them conducted for treating diseases. In this work, we will compare three approaches and also analyze their possible indications for diseases in future treatment on the basis of their distinct characteristics.
Collapse
|
8
|
Fazeli Z, Omrani MD, Ghaderian SMH. Down-regulation of nestin in mesenchymal stem cells derived from peripheral blood through blocking bone morphogenesis pathway. J Cell Commun Signal 2016; 10:273-282. [PMID: 27287702 PMCID: PMC5143318 DOI: 10.1007/s12079-016-0334-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/06/2016] [Indexed: 12/31/2022] Open
Abstract
Different signaling pathways are implicated in proliferation and differentiation of stem cells. Bone Morphogenesis Pathway (BMP) signaling was known to display an important function in osteogenic and adipogenic differentiation of mesenchymal stem cells (MSCs). In the present study, the authors investigated whether blocking BMP signaling was associated with down regulation of Nestin expression as neural stem cell marker in peripheral blood derived mesenchymal stem cells (PB-MSCs). At first, MSCs were isolated from peripheral blood by plastic adherent ability and flow cytometry analysis. After reaching the confluence, the cells were treated with medium containing Noggin as antagonist of BMP signaling upon 8 days. Real time PCR analysis indicated that the expression of Nestin was diminished in PB-MSCs by attenuating BMP signaling. The obtained results suggested that BMP signaling might have a regulatory function on the Nestin expression in mesenchymal stem cells.
Collapse
Affiliation(s)
- Zahra Fazeli
- 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
|
9
|
Ding S, Kingshott P, Thissen H, Pera M, Wang PY. Modulation of human mesenchymal and pluripotent stem cell behavior using biophysical and biochemical cues: A review. Biotechnol Bioeng 2016; 114:260-280. [DOI: 10.1002/bit.26075] [Citation(s) in RCA: 298] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 06/27/2016] [Accepted: 08/07/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Sheryl Ding
- Department of Chemistry and Biotechnology; Swinburne University of Technology; Hawthorn 3122 Victoria Australia
| | - Peter Kingshott
- Department of Chemistry and Biotechnology; Swinburne University of Technology; Hawthorn 3122 Victoria Australia
| | | | - Martin Pera
- Department of Anatomy and Neuroscience, Walter and Eliza Hall Institute of Medical Research, Florey Neuroscience and Mental Health Institute; The University of Melbourne; Victoria Australia
| | - Peng-Yuan Wang
- Department of Chemistry and Biotechnology; Swinburne University of Technology; Hawthorn 3122 Victoria Australia
- CSIRO Manufacturing; Clayton Victoria Australia
- Department of Anatomy and Neuroscience, Walter and Eliza Hall Institute of Medical Research, Florey Neuroscience and Mental Health Institute; The University of Melbourne; Victoria Australia
- Graduate Institute of Nanomedicine and Medical Engineering; College of Biomedical Engineering; Taipei Medical University; Taipei Taiwan
| |
Collapse
|
10
|
Sun W, Motta A, Shi Y, Seekamp A, Schmidt H, Gorb SN, Migliaresi C, Fuchs S. Co-culture of outgrowth endothelial cells with human mesenchymal stem cells in silk fibroin hydrogels promotes angiogenesis. ACTA ACUST UNITED AC 2016; 11:035009. [PMID: 27271291 DOI: 10.1088/1748-6041/11/3/035009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sufficient vascularization of the implant construct is required for tissue regeneration to ensure the supply of oxygen and nutrients. In our previous work, we established sonication-induced silk fibroin hydrogel to load neural stem cells for brain tissue engineering applications. In this study, we explored the application of silk fibroin as an injectable hydrogel for vascularization of soft tissues. We investigated the ability of outgrowth endothelial cells (OECs) in mono-culture or in co-culture with human bone marrow-derived mesenchymal stem cells (BM-MSCs) to form capillary networks in silk fibroin hydrogels. Furthermore, the silk fibroin hydrogel was modified with IKVAV peptide revealing a sequence derived from the extracellular matrix component laminin-1 to test its effects on angiogenesis, using unmodified and VVIAK modified silk fibroin hydrogel as controls. In monocultures of OECs, no angiogenic structures were observed in silk fibroin hydrogels. In contrast, vascular structures were abundant and increased in co-culture, as confirmed by immunocytochemistry and scanning electron microscopy (SEM) over 10 d of culture in silk fibroin-based hydrogels. Although no significant differences in angiogenic activity seem to be caused by the IKVAV peptide in our experimental settings, these results indicate that sonication-induced silk fibroin-based hydrogels support the formation of functional endothelial tubes and vascularization networks in the presence of mesenchymal cells supporting the vascular sprouting of endothelial cells.
Collapse
Affiliation(s)
- Wei Sun
- Department of Industrial Engineering and Biotech Research Center, University of Trento, via Mesiano 77, 38123 Trento, Italy. European Institute of Excellence on Tissue Engineering and Regenerative medicine, Trento, Italy
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Shen Y, Huang J, Liu L, Xu X, Han C, Zhang G, Jiang H, Li J, Lin Z, Xiong N, Wang T. A Compendium of Preparation and Application of Stem Cells in Parkinson's Disease: Current Status and Future Prospects. Front Aging Neurosci 2016; 8:117. [PMID: 27303288 PMCID: PMC4885841 DOI: 10.3389/fnagi.2016.00117] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 05/09/2016] [Indexed: 12/22/2022] Open
Abstract
Parkinson's Disease (PD) is a progressively neurodegenerative disorder, implicitly characterized by a stepwise loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) and explicitly marked by bradykinesia, rigidity, resting tremor and postural instability. Currently, therapeutic approaches available are mainly palliative strategies, including L-3,4-dihydroxy-phenylalanine (L-DOPA) replacement therapy, DA receptor agonist and deep brain stimulation (DBS) procedures. As the disease proceeds, however, the pharmacotherapeutic efficacy is inevitably worn off, worse still, implicated by side effects of motor response oscillations as well as L-DOPA induced dyskinesia (LID). Therefore, the frustrating status above has propeled the shift to cell replacement therapy (CRT), a promising restorative therapy intending to secure a long-lasting relief of patients' symptoms. By far, stem cell lines of multifarious origins have been established, which can be further categorized into embryonic stem cells (ESCs), neural stem cells (NSCs), induced neural stem cells (iNSCs), mesenchymal stem cells (MSCs), and induced pluripotent stem cells (iPSCs). In this review, we intend to present a compendium of preparation and application of multifarious stem cells, especially in relation to PD research and therapy. In addition, the current status, potential challenges and future prospects for practical CRT in PD patients will be elaborated as well.
Collapse
Affiliation(s)
- Yan Shen
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jinsha Huang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Ling Liu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Xiaoyun Xu
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Chao Han
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Guoxin Zhang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Haiyang Jiang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Jie Li
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Zhicheng Lin
- Department of Psychiatry, Harvard Medical School, Division of Alcohol and Drug Abuse, and Mailman Neuroscience Research Center, McLean Hospital Belmont, MA, USA
| | - Nian Xiong
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| | - Tao Wang
- Department of Neurology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology Wuhan, China
| |
Collapse
|
12
|
Abstract
The utilization of mesenchymal stem cells (also known as mesenchymal stromal cells, or MSCs) as a cell-based therapy for diseases that have ongoing inflammatory damage has become increasingly available. Our understanding of the cell biology of MSCs is still incomplete. However, as a result of increasing numbers of pre-clinical and clinical studies, general themes are emerging. The capacity of MSCs to reduce disease burden is largely associated with their ability to modulate the activity of the host immune responses rather than to contribute directly to tissue regeneration. As a result, they have significant potential in the treatment of chronic inflammatory disease regardless of the affected tissue. For example, MSC based therapies have been developed in the context of diseases as diverse as rheumatoid arthritis and multiple sclerosis. Here we discuss some of the principles that link these conditions, and the aspects of MSC biology that contribute to their use as a therapy for chronic inflammatory conditions.
Collapse
|
13
|
Onishi K, Jones DL, Riester SM, Lewallen EA, Lewallen DG, Sellon JL, Dietz AB, Qu W, van Wijnen AJ, Smith J. Human Adipose-Derived Mesenchymal Stromal/Stem Cells Remain Viable and Metabolically Active Following Needle Passage. PM R 2016; 8:844-54. [PMID: 26826615 DOI: 10.1016/j.pmrj.2016.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/10/2016] [Accepted: 01/20/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To assess the biological effects of passage through clinically relevant needles on the viability and metabolic activity of culture-expanded, human adipose tissue-derived mesenchymal stromal/stem cells (AMSCs). DESIGN Prospective observational pilot study. SETTING Academic medical center. PARTICIPANTS Patient-derived clinical-grade culture expanded AMSCs. INTERVENTIONS AMSCs were passed through syringes without a needle attached (control), with an 18-gauge (25.4-mm) needle attached and with a 30-gauge (19-mm) needle attached at a constant injection flow rate and constant cell concentrations. Each injection condition was completed in triplicate. MAIN OUTCOME MEASURES Cell number and viability, proliferative capacity, metabolic activity, and acute gene expression as measured by cell counts, mitochondrial activity, and quantitative real time reverse-transcription polymerase chain reaction on day 0 (immediately), day 1, and day 4 after injection. RESULTS AMSC viability was not significantly affected by injection, and cells proliferated normally regardless of study group. Postinjection, AMSCs robustly expressed both proliferation markers and extracellular matrix proteins. Stress-response mRNAs were markedly but transiently increased independently of needle size within the first day in culture postinjection. CONCLUSIONS Human, culture-expanded AMSCs maintain their viability, proliferative capacity, and metabolic function following passage through needles as small as 30-gauge at constant flow rates of 4 mL/min, despite an early, nonspecific stress/cytoprotective response. These initial findings suggest that culture-expanded AMSCs should tolerate the injection process during most cell-based therapeutic interventions.
Collapse
Affiliation(s)
- Kentaro Onishi
- Department of Physical Medicine & Rehabilitation, Mayo Clinic Sports Medicine Center, Mayo Clinic, Rochester, MN(∗)
| | - Dakota L Jones
- Department of Biomedical Engineering and Physiology, Mayo Graduate School, Mayo Clinic, Rochester, MN; Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(†)
| | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(‡)
| | - Eric A Lewallen
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(§)
| | - David G Lewallen
- Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(‖)
| | - Jacob L Sellon
- Department of Physical Medicine & Rehabilitation, Mayo Clinic Sports Medicine Center, Mayo Clinic, Rochester, MN(¶)
| | - Allan B Dietz
- Department of Biochemistry & Molecular Biology, Mayo Graduate School, Mayo Clinic, Rochester, MN; Department of Laboratory Medicine & Pathology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(#)
| | - Wenchun Qu
- Department of Physical Medicine & Rehabilitation, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN; Department of Anesthesiology Division of Pain Medicine, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(∗∗)
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Medical Sciences Building, Rm S3-69, Mayo Clinic, 200 1st St, SW, Rochester, MN 55905; Department of Biomedical Engineering and Physiology, Mayo Graduate School, Mayo Clinic, Rochester, MN; Department of Biochemistry & Molecular Biology, Mayo Graduate School, Mayo Clinic, Rochester, MN(††).
| | - Jay Smith
- Department of Physical Medicine & Rehabilitation, W14, Mayo Building, Mayo Clinic, 200 1st St, SW, Rochester, MN 55905; Department of Radiology, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN; Department of Anatomy, Mayo Clinic College of Medicine, Mayo Clinic, Rochester, MN(‡‡).
| |
Collapse
|
14
|
Kota DJ, Prabhakara KS, van Brummen AJ, Bedi S, Xue H, DiCarlo B, Cox CS, Olson SD. Propranolol and Mesenchymal Stromal Cells Combine to Treat Traumatic Brain Injury. Stem Cells Transl Med 2015; 5:33-44. [PMID: 26586775 DOI: 10.5966/sctm.2015-0065] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 09/14/2015] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED More than 6.5 million patients are burdened by the physical, cognitive, and psychosocial deficits associated with traumatic brain injury (TBI) in the U.S. Despite extensive efforts to develop neuroprotective therapies for this devastating disorder, there have been no successful outcomes in human clinical trials to date. Retrospective studies have shown that β-adrenergic receptor blockers, specifically propranolol, significantly decrease mortality of TBI through mechanisms not yet fully elucidated but are thought to counterbalance a hyperadrenergic state resulting from a TBI. Conversely, cellular therapies have been shown to improve long-term behavior following TBI, likely by reducing inflammation. Given the nonredundancy in their therapeutic mechanisms, we hypothesized that a combination of acute propranolol followed by mesenchymal stem cells (MSCs) isolated from human bone marrow would have additive effects in treating a rodent model of TBI. We have found that the treatments are well-tolerated individually and in combination with no adverse events. MSCs decrease BBB permeability at 96 hours after injury, inhibit a significant accumulation of activated microglia/macrophage in the thalamic region of the brain both short and long term, and enhance neurogenesis short term. Propranolol decreases edema and reduces the number of fully activated microglia at 7 days and the number of semiactivated microglia at 120 days. Combinatory treatment improved cognitive and memory functions 120 days following TBI. Therefore, the results here suggest a new, efficacious sequential treatment for TBI may be achieved using the β-blocker propranolol followed by MSC treatment. SIGNIFICANCE Despite continuous efforts, traumatic brain injury (TBI) remains the leading cause of death and disability worldwide in patients under the age of 44. In this study, an animal model of moderate-severe TBI was treated with an acute dose of propranolol followed by a delayed dose of human mesenchymal stem cells (MSCs), resulting in improved short- and long-term measurements. These results have direct translational application. They reinforce the inevitable clinical trial of MSCs to treat TBI by demonstrating, among other benefits, a notable decrease in chronic neuroinflammation. More importantly, these results demonstrate that MSCs and propranolol, which is increasingly being used clinically for TBI, are compatible treatments that improve overall outcome.
Collapse
Affiliation(s)
- Daniel J Kota
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Karthik S Prabhakara
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Alexandra J van Brummen
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Supinder Bedi
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Hasen Xue
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Bryan DiCarlo
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Charles S Cox
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Scott D Olson
- Department of Pediatric Surgery, University of Texas Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
15
|
|
16
|
Yang K, Park HJ, Han S, Lee J, Ko E, Kim J, Lee JS, Yu JH, Song KY, Cheong E, Cho SR, Chung S, Cho SW. Recapitulation of in vivo-like paracrine signals of human mesenchymal stem cells for functional neuronal differentiation of human neural stem cells in a 3D microfluidic system. Biomaterials 2015; 63:177-88. [PMID: 26113074 DOI: 10.1016/j.biomaterials.2015.06.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 06/10/2015] [Accepted: 06/11/2015] [Indexed: 12/22/2022]
Abstract
Paracrine signals produced from stem cells influence tissue regeneration by inducing the differentiation of endogenous stem or progenitor cells. However, many recent studies that have investigated paracrine signaling of stem cells have relied on either two-dimensional transwell systems or conditioned medium culture, neither of which provide optimal culture microenvironments for elucidating the effects of paracrine signals in vivo. In this study, we recapitulated in vivo-like paracrine signaling of human mesenchymal stem cells (hMSCs) to enhance functional neuronal differentiation of human neural stem cells (hNSCs) in three-dimensional (3D) extracellular matrices (ECMs) within a microfluidic array platform. In order to amplify paracrine signaling, hMSCs were genetically engineered using cationic polymer nanoparticles to overexpress glial cell-derived neurotrophic factor (GDNF). hNSCs were cultured in 3D ECM hydrogel used to fill central channels of the microfluidic device, while GDNF-overexpressing hMSCs (GDNF-hMSCs) were cultured in channels located on both sides of the central channel. This setup allowed for mimicking of paracrine signaling between genetically engineered hMSCs and endogenous hNSCs in the brain. Co-culture of hNSCs with GDNF-hMSCs in the 3D microfluidic system yielded reduced glial differentiation of hNSCs while significantly enhancing differentiation into neuronal cells including dopaminergic neurons. Neuronal cells produced from hNSCs differentiating in the presence of GDNF-hMSCs exhibited functional neuron-like electrophysiological features. The enhanced paracrine ability of GDNF-hMSCs was finally confirmed using an animal model of hypoxic-ischemic brain injury. This study demonstrates the presented 3D microfluidic array device can provide an efficient co-culture platform and provide an environment for paracrine signals from transplanted stem cells to control endogenous neuronal behaviors in vivo.
Collapse
Affiliation(s)
- Kisuk Yang
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Department of Biomaterials Science and Engineering, Yonsei University, Seoul 120-749, Republic of Korea
| | - Hyun-Ji Park
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Sewoon Han
- School of Mechanical Engineering, Korea University, Seoul 136-713, Republic of Korea
| | - Joan Lee
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Eunkyung Ko
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Jin Kim
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Jong Seung Lee
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Ji Hea Yu
- Department and Research Institute of Rehabilitation Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Ki Yeong Song
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Eunji Cheong
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea
| | - Seok Chung
- School of Mechanical Engineering, Korea University, Seoul 136-713, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea; Department of Neurosurgery, Yonsei University College of Medicine, Seoul 120-752, Republic of Korea.
| |
Collapse
|
17
|
Haragopal H, Yu D, Zeng X, Kim SW, Han IB, Ropper AE, Anderson JE, Teng YD. Stemness enhancement of human neural stem cells following bone marrow MSC coculture. Cell Transplant 2015; 24:645-59. [PMID: 25719952 DOI: 10.3727/096368915x687561] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Rapid loss of stemness capacity in purified prototype neural stem cells (NSCs) remains a serious challenge to basic and clinical studies aiming to repair the central nervous system. Based on the essential role of mesodermal guidance in the process of neurulation, we hypothesized that coculture of human NSCs (hNSCs) with human bone marrow-derived mesenchymal stromal stem cells (hMSCs) could enhance the stemness of hNSCs through Notch-1 signaling. We have now tested the hypothesis by assessing behaviors of hNSCs and hMSCs under systematically designed coculture conditions relative to monocultures, with or without Notch-1 manipulation in vitro. Our data demonstrate that expression levels of Notch-1 and Hes-1 as determined by immunocytochemistry are significantly higher in hNSCs cocultured with hMSCs than those of controls. Furthermore, coculturing significantly increases immunoreactivity of CD15, a neural stemness marker, but decreases CD24, a marker of neural/neuronal commitment in hNSCs. The effect is independent from the physical status of cell growth since coculture and notch signaling actually promotes hNSC adhesion. Importantly, coculture with hMSCs markedly augments hNSC proliferation rate (e.g., higher yield in G2/M phase subpopulation in a notch-dependent manner detected by flow cytometry) without diminishing their lineage differentiation capabilities. The results suggest that coculture of hNSCs with hMSCs enhances stemness biology of hNSCs partially via activation of Notch-1 signal transduction. Our finding sheds new light on mesoderm-ectoderm cell fate determination via contact-based hMSC-hNSC interactions and provides mechanistic leads for devising effective regimens to sustain and augment stemness of in vitro established hNSC and hMSC lines for basic science, translational and clinical applications.
Collapse
Affiliation(s)
- Hariprakash Haragopal
- Department of Neurosurgery, Harvard Medical School and the Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Matsiko A, Gleeson JP, O'Brien FJ. Scaffold Mean Pore Size Influences Mesenchymal Stem Cell Chondrogenic Differentiation and Matrix Deposition. Tissue Eng Part A 2015; 21:486-97. [DOI: 10.1089/ten.tea.2013.0545] [Citation(s) in RCA: 163] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Amos Matsiko
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Trinity Center for Bioengineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, Dublin, Ireland
| | - John P. Gleeson
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Trinity Center for Bioengineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, Dublin, Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Trinity Center for Bioengineering, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, Dublin, Ireland
| |
Collapse
|
19
|
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
|
20
|
Huang J, Sha H, Wang G, Bao G, Lu S, Luo Q, Tan Q. Isolation and characterization of ex vivo expanded mesenchymal stem cells obtained from a surgical patient. Mol Med Rep 2014; 11:1777-83. [PMID: 25376882 PMCID: PMC4270342 DOI: 10.3892/mmr.2014.2892] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 09/29/2014] [Indexed: 12/20/2022] Open
Abstract
The aim of the present study was to investigate the morphological characteristics and pluripotent differentiation potential of human bone marrow mesenchymal stem cells (hBMMSCs) in vitro and in vivo. Bone marrow cells were isolated from a rib fragment of an adult surgical patient, hBMMSCs were isolated based on plastic adherence and expanded ex vivo and phenotyping was performed. Pluripotent differentiation assays for adipogenesis, myogenesis and osteogenesis were conducted. Hematopoietic reconstruction of sublethally irradiated nude mice was performed by infusion of hBMMSCs. The gene expression profiles of early and late hBMMSCs were examined. The rate of CD31-positive cells was 31.1% in passage (P)4 hBMMSCs and 18.6% in P10 hBMMSCs. CD105 and CD106 were expressed in 99 and 95% of P25 hBMMSCs, respectively. Lipid droplets appeared at day 18 post induction. For osteogenesis, palpable masses were grossly observed from day 35 post inoculation of hBMMSCs. Hematoxylin and eosin staining further revealed chondrocytes and bone tissues. For myogenesis, at day six post subcutaneous inoculation, hBMMSCs differentiated into myocytes and were positive for myoglobin and MyoD1. In irradiated nude mice reconstituted by hBMMSCs, the white blood cell count briefly decreased following irradiation; however, it gradually recovered. In the irradiated nude mice reconstituted with hBMMSCs, CD45- and CD34-positive cells were detected 72 h post induction. Gene microarray analysis of P7 and P57 hBMMSCs demonstrated that 20 genes were upregulated >2 fold and 40 genes were downregulated >2 fold in P57 hBMMSCs. In conclusion, the isolated HBMMSCs possessed pluripotent differentiation potential and it was feasible and safe to use hBMMSCs within 30 passages.
Collapse
Affiliation(s)
- Jia Huang
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Huifan Sha
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Guan Wang
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Guoliang Bao
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Shun Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Qingquan Luo
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| | - Qiang Tan
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, P.R. China
| |
Collapse
|
21
|
Sart S, Liu Y, Ma T, Li Y. Microenvironment Regulation of Pluripotent Stem Cell-Derived Neural Progenitor Aggregates by Human Mesenchymal Stem Cell Secretome. Tissue Eng Part A 2014; 20:2666-79. [DOI: 10.1089/ten.tea.2013.0437] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Sébastien Sart
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Yijun Liu
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Teng Ma
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, Florida
| |
Collapse
|
22
|
Skoloudik L, Chrobok V, Kalfert D, Koci Z, Filip S. Multipotent mesenchymal stromal cells in otorhinolaryngology. Med Hypotheses 2014; 82:769-73. [DOI: 10.1016/j.mehy.2014.03.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 03/02/2014] [Accepted: 03/19/2014] [Indexed: 12/19/2022]
|
23
|
Glavaski-Joksimovic A, Bohn MC. Mesenchymal stem cells and neuroregeneration in Parkinson's disease. Exp Neurol 2013; 247:25-38. [DOI: 10.1016/j.expneurol.2013.03.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/14/2013] [Indexed: 02/06/2023]
|
24
|
Mesenchymal stem cells from human umbilical cord express preferentially secreted factors related to neuroprotection, neurogenesis, and angiogenesis. PLoS One 2013; 8:e72604. [PMID: 23991127 PMCID: PMC3749979 DOI: 10.1371/journal.pone.0072604] [Citation(s) in RCA: 228] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 07/12/2013] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are promising tools for the treatment of diseases such as infarcted myocardia and strokes because of their ability to promote endogenous angiogenesis and neurogenesis via a variety of secreted factors. MSCs found in the Wharton’s jelly of the human umbilical cord are easily obtained and are capable of transplantation without rejection. We isolated MSCs from Wharton’s jelly and bone marrow (WJ-MSCs and BM-MSCs, respectively) and compared their secretomes. It was found that WJ-MSCs expressed more genes, especially secreted factors, involved in angiogenesis and neurogenesis. Functional validation showed that WJ-MSCs induced better neural differentiation and neural cell migration via a paracrine mechanism. Moreover, WJ-MSCs afforded better neuroprotection efficacy because they preferentially enhanced neuronal growth and reduced cell apoptotic death of primary cortical cells in an oxygen-glucose deprivation (OGD) culture model that mimics the acute ischemic stroke situation in humans. In terms of angiogenesis, WJ-MSCs induced better microvasculature formation and cell migration on co-cultured endothelial cells. Our results suggest that WJ-MSC, because of a unique secretome, is a better MSC source to promote in vivo neurorestoration and endothelium repair. This study provides a basis for the development of cell-based therapy and carrying out of follow-up mechanistic studies related to MSC biology.
Collapse
|
25
|
Fraga JS, Silva NA, Lourenço AS, Gonçalves V, Neves NM, Reis RL, Rodrigues AJ, Manadas B, Sousa N, Salgado AJ. Unveiling the effects of the secretome of mesenchymal progenitors from the umbilical cord in different neuronal cell populations. Biochimie 2013; 95:2297-303. [PMID: 23851197 DOI: 10.1016/j.biochi.2013.06.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Accepted: 06/19/2013] [Indexed: 12/11/2022]
Abstract
It has been previously shown that the secretome of Human Umbilical Cord Perivascular Cells (HUCPVCs), known for their mesenchymal like stem cell character, is able to increase the metabolic viability and hippocampal neuronal cell densities. However, due to the different micro-environments of the distinct brain regions it is important to study if neurons isolated from different areas have similar, or opposite, reactions when in the presence of HUCPVCs secretome (in the form of conditioned media-CM). In this work we: 1) studied how cortical and cerebellar neuronal primary cultures behaved when incubated with HUCPVCs CM and 2) characterized the differences between CM collected at two different conditioning time points. Primary cultures of cerebellar and cortical neurons were incubated with HUCPVCs CM (obtained 24 and 96 h after three days of culturing). HUCPVCs CM had a higher impact on the metabolic viability and proliferation of cortical cultures, than the cerebellar ones. Regarding neuronal cell densities it was observed that with 24 h CM condition there were higher number MAP-2 positive cells, a marker for fully differentiated neurons; this was, once again, more evident in cortical cultures. In an attempt to characterize the differences between the two conditioning time points a proteomics approach was followed, based on 2D Gel analysis followed by the identification of selected spots by tandem mass spectrometry. Results revealed important differences in proteins that have been previously related with phenomena such as neurl cell viability, proliferation and differentiation, namely 14-3-3, UCHL1, hsp70 and peroxiredoxin-6. In summary, we demonstrated differences on how neurons isolated from different brain regions react to HUCPVCs secretome and we have identified different proteins (14-3-3 and hsp70) in HUCPVCs CM that may explain the above-referred results.
Collapse
Affiliation(s)
- J S Fraga
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Lin CH, Lee HT, Lee SD, Lee W, Cho CWC, Lin SZ, Wang HJ, Okano H, Su CY, Yu YL, Hsu CY, Shyu WC. Role of HIF-1α-activated Epac1 on HSC-mediated neuroplasticity in stroke model. Neurobiol Dis 2013; 58:76-91. [PMID: 23702312 DOI: 10.1016/j.nbd.2013.05.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Revised: 05/02/2013] [Accepted: 05/10/2013] [Indexed: 12/27/2022] Open
Abstract
Exchange protein activated by cAMP-1 (Epac1) plays an important role in cell proliferation, cell survival and neuronal signaling, and activation of Epac1 in endothelial progenitor cells increases their homing to ischemic muscles and promotes neovascularization in a model of hind limb ischemia. Moreover, upregulation of Epac1 occurs during organ development and in diseases such as myocardial hypertrophy, diabetes, and Alzheimer's disease. We report here that hypoxia upregulated Epac1 through HIF-1α induction in the CD34-immunosorted human umbilical cord blood hematopoietic stem cells (hUCB(34)). Importantly, implantation of hUCB(34) subjected to hypoxia-preconditioning (HP-hUCB(34)) improved stroke outcome, more than did implantation of untreated hUCB(34), in rodents subjected to cerebral ischemia, and this required Epac1-to-matrix metalloprotease (MMP) signaling. This improved therapeutic efficacy correlated with better engraftment and differentiation of these cells in the ischemic host brain. In addition, more than did implantation of untreated HP-hUCB(34), implantation of HP-hUCB(34) improved cerebral blood flow into the ischemic brain via induction of angiogenesis, facilitated proliferation/recruitment of endogenous neural progenitor cells in the ischemic brain, and promoted neurite outgrowth following cerebral ischemia. Consistent with our proposed role of Epac1-to-MMP signaling in hypoxia-preconditioning, the above mentioned effects of implanting HP-hUCB(34) could be abolished by pharmacological inhibition and genetic disruption/deletion of Epac1 or MMPs. We have discovered a HIF-1α-to-Epac1-to-MMP signaling pathway that is required for the improved therapeutic efficacy resulting from hypoxia preconditioning of hUCB(34) in vitro prior to their implantation into the host brain in vivo.
Collapse
Affiliation(s)
- Chen-Huan Lin
- Department of Neurology, China Medical University Hospital, Taichung, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Characterization of in vitro cultured bone marrow and adipose tissue-derived mesenchymal stem cells and their ability to express neurotrophic factors. Cell Biol Int 2013; 36:1239-49. [PMID: 22994924 DOI: 10.1042/cbi20110618] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
MSCs (mesenchymal stem cells) have attracted attention as a promising tool for regenerative medicine and transplantation therapy. MSCs exert neuroprotective effects by secreting a number of factors in vitro and in vivo. Similar characteristics are found in ADSCs (adipose-derived stem cells) and BMSCs (bone marrow stromal cells). Multipotent capability, easy accessibility and rapid proliferation of ADSCs have been established. Our main objective was to compare cell viability, growth rate, expression of neurotrophic factors and nestin genes in ADSCs and BMSCs. Cell doubling time and proliferation rate indicate that ADSCs has a higher proliferation rate than BMSCs. ADSCs and BMSCs express a similar pattern of CD71 and CD90 markers. Nestin immunostaining showed that ADSCs and BMSCs are immunopositive. The expression of neurotrophic factors genes in ADSCs proved similar to that of BMSCs genes. Thus adipose tissue stem cells with a high proliferation rate can express nestin and neurotrophic factor genes. Therefore ADSCs may be useful in future cell replacement therapies and help improve neurodegenerative diseases.
Collapse
|
28
|
da Cruz e Alves-de-Moraes LB, Ribeiro-Paes JT, Longo BM, Ferrazoli EG, de Andrade TGCS. Effect of the bone marrow cell transplantation on elevated plus-maze performance in hippocampal-injured mice. Behav Brain Res 2013; 248:32-40. [PMID: 23578758 DOI: 10.1016/j.bbr.2013.03.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 03/25/2013] [Accepted: 03/31/2013] [Indexed: 11/15/2022]
Abstract
Several reports have shown that the hippocampus plays an important role in different aspects of the emotional control. There is evidence that lesions in this structure cause behavioral disinhibition, with reduction of reactions expressing fear and anxiety. Thus, to portray the aptitude of cell therapy to abrogate injuries of hippocampal tissue, we examined the behavioral effects of bone marrow mononuclear cells (BMMCs) transplantation on C57BL/6 mice that had the hippocampus damaged by electrolytic lesion. For this purpose, mice received, seven days after bilateral electrolytic lesion in the dorsal hippocampus, culture medium or BMMCs expressing the enhanced green fluorescent protein (EGFP) transgene. One week after transplantation, animals were tested in the elevated plus-maze (EPM). On the whole, three assessment sessions in the EPM were carried out, with seven days separating each trial. Thirty-five days after the induction of injury, mice were sacrificed and their brains removed for immunohistochemistry. The behavioral evaluation showed that the hippocampal lesion caused disinhibition, an effect which was slightly lessened, from the second EPM test, in transplanted subjects. On the other hand, immunohistochemical data revealed an insignificant presence of EGFP(+) cells inside the brains of injured mice. In view of such scenario, we hypothesized that the subtle rehabilitation of the altered behavior might be a result from a paracrine effect from the transplanted cells. This might have been caused by the release of bioactive factors capable of boosting endogenous recuperative mechanisms for a partial regaining of the hippocampal functions.
Collapse
|
29
|
Aizman I, McGrogan M, Case CC. Quantitative microplate assay for studying mesenchymal stromal cell-induced neuropoiesis. Stem Cells Transl Med 2013; 2:223-32. [PMID: 23430693 DOI: 10.5966/sctm.2012-0119] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Transplanting mesenchymal stromal cells (MSCs) or their derivatives in a neurodegenerative environment is believed to be beneficial because of the trophic support, migratory guidance, and neurogenic stimuli they provide. There is a growing need for in vitro models of mesenchymal-neural cell interactions to enable identification of mediators of the MSC activity and quantitative assessment of neuropoietic potency of MSC preparations. Here, we characterize a microplate-format coculture system in which primary embryonic rat cortex cells are directly cocultured with human MSCs on cell-derived extracellular matrix (ECM) in the absence of exogenous growth factors. In this system, expression levels of the rat neural stem/early progenitor marker nestin, as well as neuronal and astrocytic markers, directly depended on MSC dose, whereas an oligodendrogenic marker exhibited a biphasic MSC-dose response, as measured using species-specific quantitative reverse transcription-polymerase chain reaction in total cell lysates and confirmed using immunostaining. Both neural cell proliferation and differentiation contributed to the MSC-mediated neuropoiesis. ECM's heparan sulfate proteoglycans were essential for the growth of the nestin-positive cell population. Neutralization studies showed that MSC-derived fibroblast growth factor 2 was a major and diffusible inducer of rat nestin, whereas MSC-derived bone morphogenetic proteins (BMPs), particularly, BMP4, were astrogenesis mediators, predominantly acting in a coculture setting. This system enables analysis of multifactorial MSC-neural cell interactions and can be used for elucidating the neuropoietic potency of MSCs and their derivative preparations.
Collapse
|
30
|
Wang Y, Tu W, Lou Y, Xie A, Lai X, Guo F, Deng Z. Mesenchymal stem cells regulate the proliferation and differentiation of neural stem cells through Notch signaling. Cell Biol Int 2013; 33:1173-9. [DOI: 10.1016/j.cellbi.2009.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 05/30/2009] [Accepted: 08/17/2009] [Indexed: 01/22/2023]
|
31
|
Cell Sources for Tissue Engineering: Mesenchymal Stem Cells. Biomater Sci 2013. [DOI: 10.1016/b978-0-08-087780-8.00111-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
32
|
Nery AA, Nascimento IC, Glaser T, Bassaneze V, Krieger JE, Ulrich H. Human mesenchymal stem cells: from immunophenotyping by flow cytometry to clinical applications. Cytometry A 2012; 83:48-61. [PMID: 23027703 DOI: 10.1002/cyto.a.22205] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 08/15/2012] [Accepted: 08/22/2012] [Indexed: 12/25/2022]
Abstract
Modern medicine will unequivocally include regenerative medicine as a major breakthrough in the re-establishment of damaged or lost tissues due to degenerative diseases or injury. In this scenario, millions of patients worldwide can have their quality of life improved by stem cell implantation coupled with endogenous secretion or administration of survival and differentiation promoting factors. Large efforts, relying mostly on flow cytometry and imaging techniques, have been put into cell isolation, immunophenotyping, and studies of differentiation properties of stem cells of diverse origins. Mesenchymal stem cells (MSCs) are particularly relevant for therapy due to their simplicity of isolation. A minimal phenotypic pattern for the identification of MSCs cells requires them to be immunopositive for CD73, CD90, and CD105 expression, while being negative for CD34, CD45, and HLA-DR and other surface markers. MSCs identified by their cell surface marker expression pattern can be readily purified from patient's bone marrow and adipose tissues. Following expansion and/or predifferentiation into a desired tissue type, stem cells can be reimplanted for tissue repair in the same patient, virtually eliminating rejection problems. Transplantation of MSCs is subject of almost 200 clinical trials to cure and treat a very broad range of conditions, including bone, heart, and neurodegenerative diseases. Immediate or medium term improvements of clinical symptoms have been reported as results of many clinical studies.
Collapse
Affiliation(s)
- Arthur A Nery
- Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
33
|
Hepatocyte growth factor mediates mesenchymal stem cell–induced recovery in multiple sclerosis models. Nat Neurosci 2012; 15:862-70. [PMID: 22610068 PMCID: PMC3427471 DOI: 10.1038/nn.3109] [Citation(s) in RCA: 330] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 04/17/2012] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells have emerged as a potential therapy for a range of neural insults. In animal models of multiple sclerosis, an autoimmune disease that targets oligodendrocytes and myelin, treatment with human MSCs results in functional improvement that reflects both modulation of the immune response and myelin repair. Here we demonstrate that conditioned medium (CM) from human MSCs reduces functional deficits in mouse MOG35–55-induced EAE and promotes the development of oligodendrocytes and neurons. Functional assays identify a critical role for Hepatocyte Growth Factor (HGF) and its primary receptor cMet in MSCs stimulated recovery in EAE, neural cell development and remyelination. Active MSC-CM contains HGF and exogenously supplied HGF promotes recovery in EAE while cMet and anti-HGF antibodies block the functional recovery mediated by HGF and MSC-CM. Systemic treatment with HGF dramatically accelerated remyelination in lysolecithin-induced rat dorsal spinal cord lesions and in slice cultures. Together these data strongly implicate HGF in mediating MSC-stimulated functional recovery in animal models of multiple sclerosis.
Collapse
|
34
|
Murine bone marrow stromal cells pulsed with homologous tumor-derived exosomes inhibit proliferation of liver cancer cells. Clin Transl Oncol 2012; 14:764-73. [PMID: 22855153 DOI: 10.1007/s12094-012-0860-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 12/20/2011] [Indexed: 12/17/2022]
Abstract
BACKGROUND Increasing evidence shows that bone marrow stromal cells (BMSCs) have antitumor activities both in vitro and in animal models. Further studies fleshed out the supportive data that the antitumor activity of BMSCs could be markedly enhanced by cytokines such as IL-2 and IFN-β (interferon). However, powerful strategies to activate BMSCs other than by genetically engineering interventions are still required. METHODS In this study, new methods of generating antitumor activities of murine marrow-originated MSCs pulsed with homologous tumor-derived exosomes (TEX) were explored to yield potent immune effectors against hepatocellular carcinoma cells in vitro. RESULTS The results showed that BMSCs pulsed with exosomes and IFN-γ exhibited increased migration ability with a result of 163.22 ± 26.90 versus 129.89 ± 29.28 cells/HP by transwell determination (p < 0.05). The inhibition of homologous hepatocellular carcinoma cells line H(22) cells by exosomes pulsed BMSCs was significantly increased by 41.9 % compared with control (p < 0.05), and flow cytometry analysis showed that the cell cycle of H(22) cells was arrested in G(0)/G(1) phase. Meanwhile, western blot analysis showed that PCNA protein expression in the supernatant of H(22) cells was significantly decreased. CONCLUSIONS This study demonstrated that BMSCs pulsed with TEX could enhance its antitumor activities, which might be regarded as a novel promising antitumor treatment.
Collapse
|
35
|
The immunomodulatory and neuroprotective effects of mesenchymal stem cells (MSCs) in experimental autoimmune encephalomyelitis (EAE): a model of multiple sclerosis (MS). Int J Mol Sci 2012; 13:9298-9331. [PMID: 22942767 PMCID: PMC3430298 DOI: 10.3390/ijms13079298] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 07/11/2012] [Accepted: 07/11/2012] [Indexed: 02/07/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent cells that differentiate into the mesenchymal lineages of adipocytes, osteocytes and chondrocytes. MSCs can also transdifferentiate and thereby cross lineage barriers, differentiating for example into neurons under certain experimental conditions. MSCs have anti-proliferative, anti-inflammatory and anti-apoptotic effects on neurons. Therefore, MSCs were tested in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), for their effectiveness in modulating the pathogenic process in EAE to develop effective therapies for MS. The data in the literature have shown that MSCs can inhibit the functions of autoreactive T cells in EAE and that this immunomodulation can be neuroprotective. In addition, MSCs can rescue neural cells via a mechanism that is mediated by soluble factors, which provide a suitable environment for neuron regeneration, remyelination and cerebral blood flow improvement. In this review, we discuss the effectiveness of MSCs in modulating the immunopathogenic process and in providing neuroprotection in EAE.
Collapse
|
36
|
Harris VK, Faroqui R, Vyshkina T, Sadiq SA. Characterization of autologous mesenchymal stem cell-derived neural progenitors as a feasible source of stem cells for central nervous system applications in multiple sclerosis. Stem Cells Transl Med 2012. [PMID: 23197858 DOI: 10.5966/sctm.2012-0015] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Bone marrow mesenchymal stem cell-derived neural progenitors (MSC-NPs) are a potential therapeutic source of cells that have been shown to be efficacious in a preclinical model of multiple sclerosis (MS). To examine the feasibility of using MSC-NPs as an autologous source of cells to promote central nervous system (CNS) repair in MS, this study characterized human MSC-NPs from a panel of both MS and non-MS donors. Expanded MSCs showed similar characteristics in terms of growth and cell surface phenotype, regardless of the donor disease status. MSC-NPs derived from all MSCs showed a consistent pattern of gene expression changes that correlated with neural commitment and increased homogeneity. Furthermore, the reduced expression of mesodermal markers and reduced capacity for adipogenic or osteogenic differentiation in MSC-NPs compared with MSCs suggested that MSC-NPs have reduced potential of unwanted mesodermal differentiation upon CNS transplantation. The immunoregulatory function of MSC-NPs was similar to that of MSCs in their ability to suppress T-cell proliferation and to promote expansion of FoxP3-positive T regulatory cells in vitro. In addition, MSC-NPs promoted oligodendroglial differentiation from brain-derived neural stem cells that correlated with the secretion of bioactive factors. Our results provide a set of identity characteristics for autologous MSC-NPs and suggest that the in vitro immunoregulatory and trophic properties of these cells may have therapeutic value in the treatment of MS.
Collapse
Affiliation(s)
- Violaine K Harris
- Multiple Sclerosis Research Center of New York, New York, New York 10019, USA
| | | | | | | |
Collapse
|
37
|
Habib HS, Halawa TF, Atta HM. Therapeutic applications of mesenchymal stroma cells in pediatric diseases: current aspects and future perspectives. Med Sci Monit 2012; 17:RA233-239. [PMID: 22037754 PMCID: PMC3539490 DOI: 10.12659/msm.882036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells or stroma cells (MSCs) were recently proven to play various therapeutic roles when used in clinical trials to control various inflammatory, neoplastic and immunologic diseases in children. Clinical trials show some promising results, particularly in diseases where conventional therapy is still ineffective. However, experimental studies sometimes show conflicting results. This review aims to assess the current therapeutic role of MSCs in the control of several pediatric diseases and elaborate on their future applications by reviewing published studies. A review of published studies on this subject based on Pubmed and Medical Subject Heading databases, with search for all relevant articles focusing on results of clinical trials to evaluate the clinical applications of MSCs. The review includes documentation of positive as well as negative applications of MSCs focused on pediatric diseases. MSCs have important immunosuppressive and antifibrotic effects that need to be employed to help patients with diseases for which no conventional management has proven to be effective. They may be also be used as an adjuvant to conventional therapeutic modalities to consolidate recovery. This review sheds light on the significance of the use of MSCs for the treatment of various pediatric diseases and focuses on promising applications. Most of the reported studies agree about the favorable use of MSCs in various diseases; however, more clinical trials, involving larger numbers of patients, need to be conducted in order to refine the outcome of the therapeutic methods and establish standardized protocols.
Collapse
Affiliation(s)
- Hamid S Habib
- Department of Pediatrics, King Abdulaziz University, Jeddah, Rabigh Branch, Saudi Arabia
| | | | | |
Collapse
|
38
|
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
|
39
|
Connick P, Kolappan M, Crawley C, Webber DJ, Patani R, Michell AW, Du MQ, Luan SL, Altmann DR, Thompson AJ, Compston A, Scott MA, Miller DH, Chandran S. Autologous mesenchymal stem cells for the treatment of secondary progressive multiple sclerosis: an open-label phase 2a proof-of-concept study. Lancet Neurol 2012; 11:150-6. [PMID: 22236384 PMCID: PMC3279697 DOI: 10.1016/s1474-4422(11)70305-2] [Citation(s) in RCA: 465] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background More than half of patients with multiple sclerosis have progressive disease characterised by accumulating disability. The absence of treatments for progressive multiple sclerosis represents a major unmet clinical need. On the basis of evidence that mesenchymal stem cells have a beneficial effect in acute and chronic animal models of multiple sclerosis, we aimed to assess the safety and efficacy of these cells as a potential neuroprotective treatment for secondary progressive multiple sclerosis. Methods Patients with secondary progressive multiple sclerosis involving the visual pathways (expanded disability status score 5·5–6·5) were recruited from the East Anglia and north London regions of the UK. Participants received intravenous infusion of autologous bone-marrow-derived mesenchymal stem cells in this open-label study. Our primary objective was to assess feasibility and safety; we compared adverse events from up to 20 months before treatment until up to 10 months after the infusion. As a secondary objective, we chose efficacy outcomes to assess the anterior visual pathway as a model of wider disease. Masked endpoint analyses was used for electrophysiological and selected imaging outcomes. We used piecewise linear mixed models to assess the change in gradients over time at the point of intervention. This trial is registered with ClinicalTrials.gov, number NCT00395200. Findings We isolated, expanded, characterised, and administered mesenchymal stem cells in ten patients. The mean dose was 1·6×106 cells per kg bodyweight (range 1·1–2·0). One patient developed a transient rash shortly after treatment; two patients had self-limiting bacterial infections 3–4 weeks after treatment. We did not identify any serious adverse events. We noted improvement after treatment in visual acuity (difference in monthly rates of change −0·02 logMAR units, 95% CI −0·03 to −0·01; p=0·003) and visual evoked response latency (−1·33 ms, −2·44 to −0·21; p=0·020), with an increase in optic nerve area (difference in monthly rates of change 0·13 mm2, 0·04 to 0·22; p=0·006). We did not identify any significant effects on colour vision, visual fields, macular volume, retinal nerve fibre layer thickness, or optic nerve magnetisation transfer ratio. Interpretation Autologous mesenchymal stem cells were safely given to patients with secondary progressive multiple sclerosis in our study. The evidence of structural, functional, and physiological improvement after treatment in some visual endpoints is suggestive of neuroprotection. Funding Medical Research Council, Multiple Sclerosis Society of Great Britain and Northern Ireland, Evelyn Trust, NHS National Institute for Health Research, Cambridge and UCLH Biomedical Research Centres, Wellcome Trust, Raymond and Beverly Sackler Foundation, and Sir David and Isobel Walker Trust.
Collapse
Affiliation(s)
- Peter Connick
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Neuroprotective effects of human mesenchymal stem cells on neural cultures exposed to 6-hydroxydopamine: implications for reparative therapy in Parkinson’s disease. Apoptosis 2011; 17:289-304. [DOI: 10.1007/s10495-011-0679-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
41
|
Abstract
Multiple sclerosis (MS) is a common neurological disease and a major cause of disability, particularly affecting young adults. It is characterized by patches of damage occurring throughout the brain and spinal cord, with loss of myelin sheaths - the insulating material around nerve fibres that allows normal conduction of nerve impulses - accompanied by loss of cells that make myelin (oligodendrocytes). In addition, we now know that there is damage to nerve cells (neurones) and their fibres (axons) too, and that this occurs both within these discrete patches and in tissue between them. The cause of MS remains unknown, but an autoimmune reaction against oligodendrocytes and myelin is generally assumed to play a major role, and early acute MS lesions almost invariably show prominent inflammation. Efforts to develop cell therapy in MS have long been directed towards directly implanting cells capable of replacing lost oligodendrocytes and regenerating myelin sheaths. Accordingly, the advent of techniques to generate large numbers of oligodendrocytes from embryonic stem cells appeared a significant step towards new stem cell treatments for MS; while the emerging consensus that adult stem cells from, for example, the bone marrow had far less potential to turn into oligodendrocytes was thought to cast doubt on their potential value in this disease. A number of scientific and medical concerns, not least the risk of tumour formation associated with embryonic stem cells, have however, prevented any possible clinical testing of these cells in patients. More recently, increasing understanding of the complexity of tissue damage in MS has emphasized that successful cell therapy may need to achieve far more than simply offering a source of replacement myelin-forming cells. The many and varied reparative properties of bone marrow-derived (mesenchymal) stem cells may well offer new and attractive possibilities for developing cell-based treatments for this difficult and disabling condition.
Collapse
Affiliation(s)
- N Scolding
- Department of Neurology, Frenchay Hospital, University of Bristol, UK.
| |
Collapse
|
42
|
Cristofanilli M, Harris VK, Zigelbaum A, Goossens AM, Lu A, Rosenthal H, Sadiq SA. Mesenchymal stem cells enhance the engraftment and myelinating ability of allogeneic oligodendrocyte progenitors in dysmyelinated mice. Stem Cells Dev 2011; 20:2065-76. [PMID: 21299379 DOI: 10.1089/scd.2010.0547] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Multiple sclerosis is an autoimmune disease characterized by demyelination and axonal loss throughout the central nervous system. No regenerative treatment exists for patients who fail to respond to conventional immunosuppressive and immunomodulating drugs. In this scenario, stem cell therapy poses as a rational approach for neurological regeneration. Transplantation of embryonic-derived oligodendrocyte progenitor cells (OPCs) has been shown to promote remyelination and ameliorate animal models of neurodegenerative diseases. However, its therapeutic application is limited due to potential transplant rejection. In multiple sclerosis, an added concern is that transplant rejection would be most pronounced at sites of previous lesions, exacerbating a hyperactive immune response which could prevent remyelination and precipitate additional demyelination. Routine systemic immunosuppression may not be sufficient to prevent transplant rejection-associated immune reactions in the cerebral microenvironment. Mesenchymal stem cells (MSCs), due to their homing properties and inherent immunosuppressive nature, are a promising tool for clinical application targeted toward immunosuppression at sites of injury. In this study, we used a co-transplantation strategy to investigate the effect of syngeneic MSCs on the survival and remyelination abilities of allogeneic OPCs in adult nonimmunosuppressed shiverer mice. At all time points examined, cotransplantation with MSCs increased OPC engraftment, migration, and maturation in myelinating oligodendrocytes, which produced widespread myelination in the host corpus callosum. In addition, MSCs reduced microglia activation and astrocytosis in the brain of transplanted animals as well as T-cell proliferation in vitro. These data suggest that combining the immunomodulatory and trophic properties of MSCs with the myelinating ability of OPCs might be a suitable strategy for promoting neurological regeneration in demyelinating diseases.
Collapse
|
43
|
Robinson AP, Foraker JE, Ylostalo J, Prockop DJ. Human stem/progenitor cells from bone marrow enhance glial differentiation of rat neural stem cells: a role for transforming growth factor β and Notch signaling. Stem Cells Dev 2011; 20:289-300. [PMID: 20575640 PMCID: PMC3128772 DOI: 10.1089/scd.2009.0444] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 06/23/2010] [Indexed: 12/20/2022] Open
Abstract
Multipotent stem/progenitor cells from bone marrow stroma (mesenchymal stromal cells or MSCs) were previously shown to enhance proliferation and differentiation of neural stem cells (NSCs) in vivo, but the molecular basis of the effect was not defined. Here coculturing human MSCs (hMSCs) with rat NSCs (rNSCs) was found to stimulate astrocyte and oligodendrocyte differentiation of the rNSCs. To survey the signaling pathways involved, RNA from the cocultures was analyzed by species-specific microarrays. In the hMSCs, there was an upregulation of transcripts for several secreted factors linked to differentiation: bone morphogenetic protein 1 (BMP1), hepatocyte growth factor (HGF), and transforming growth factor isoforms (TGFβ1 and TGFβ3). In both the hMSCs and the rNSCs, there was an upregulation of transcripts for Notch signaling. The role of TGFβ1 was verified by the demonstration that hMSCs in coculture increased secretion of TGFβ1, the rNSCs expressed the receptor, and an inhibitor of TGFβ signaling blocked differentiation. The role of Notch signaling was verified by the demonstration that in the cocultures hMSCs expressed a Notch ligand at sites of cell contact with rNSCs, and the rNSCs expressed the receptor, Notch 1. Increased Notch signaling in both cell types was then demonstrated by assays of transcript expression and by a reporter construct for downstream targets of Notch signaling. The results demonstrated that glial differentiation of the rNSCs in the cocultures was driven by increased secretion of soluble factors such as TGFβ1 by the hMSCs and probably through increased cell contact signaling between the hMSCs and rNSCs through the Notch pathway.
Collapse
|
44
|
Ribeiro CA, Salgado AJ, Fraga JS, Silva NA, Reis RL, Sousa N. The secretome of bone marrow mesenchymal stem cells-conditioned media varies with time and drives a distinct effect on mature neurons and glial cells (primary cultures). J Tissue Eng Regen Med 2011; 5:668-72. [DOI: 10.1002/term.365] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 07/29/2010] [Indexed: 01/01/2023]
|
45
|
Shafaei H, Esmaeili A, Mardani M, Razavi S, Hashemibeni B, Nasr-Esfahani MH, Shiran MB, Esfandiari E. Effects of human placental serum on proliferation and morphology of human adipose tissue-derived stem cells. Bone Marrow Transplant 2011; 46:1464-71. [DOI: 10.1038/bmt.2010.313] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
46
|
Human mesenchymal stem cells infiltrate the spinal cord, reduce demyelination, and localize to white matter lesions in experimental autoimmune encephalomyelitis. J Neuropathol Exp Neurol 2010; 69:1087-95. [PMID: 20940628 DOI: 10.1097/nen.0b013e3181f97392] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Mesenchymal stem cells (MSCs) can abrogate the animal model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE), but whether this therapeutic effect occurs entirely through systemic immune modulation and whether CNS infiltration occurs after peripheral delivery are uncertain. We studied the clinical and neuropathologic effects of intravenously administered human MSCs (hMSCs) in C57BL/6 mice with EAE. Human MSCs significantly reduced the clinical disease severity, particularly in later disease. Large numbers of hMSCs migrated into gray and white matter at all levels of the spinal cord in both naive mice and mice with EAE. In the latter, hMSCs accumulated over time in demyelinated areas. There were 2 distinct morphological appearances of the hMSCs in the tissue, that is, rounded and less numerous process-bearing forms; very few expressed neural markers. The number of spinal cord white matter lesions and areas of white matter demyelination were reduced after hMSC treatment compared with control treatment. These findings show that central nervous system infiltration occurs after peripheral delivery of hMSCs, that they accumulate where there is myelin damage, and that they are associated with a reduced extent of demyelination. These data support a potential role for hMSCs in autologous cell therapy in multiple sclerosis.
Collapse
|
47
|
Lepski G, Jannes CE, Strauss B, Marie SK, Nikkhah G. Survival and Neuronal Differentiation of Mesenchymal Stem Cells Transplanted into the Rodent Brain Are Dependent upon Microenvironment. Tissue Eng Part A 2010; 16:2769-82. [DOI: 10.1089/ten.tea.2009.0686] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Affiliation(s)
- Guilherme Lepski
- Department of Stereotactic and Functional Neurosurgery, Albert Ludwigs University, Frieburg, Germany
| | - Cinthia E. Jannes
- Laboratory of Molecular Biology LIM15, Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Bryan Strauss
- Laboratory of Molecular Biology, INCOR, School of Medicine, University of Sao Paulo, Sao Paulo, Brazil
| | - Suely K.N. Marie
- Laboratory of Molecular Biology LIM15, Medical School, University of Sao Paulo, Sao Paulo, Brazil
| | - Guido Nikkhah
- Department of Stereotactic and Functional Neurosurgery, Neurocentre, Albert-Ludwig University, Freiburg, Germany
| |
Collapse
|
48
|
Lukaszewicz AI, McMillan MK, Kahn M. Small molecules and stem cells. Potency and lineage commitment: the new quest for the fountain of youth. J Med Chem 2010; 53:3439-53. [PMID: 20047330 DOI: 10.1021/jm901361d] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Agnès I Lukaszewicz
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Los Angeles, CA 90033, USA
| | | | | |
Collapse
|
49
|
Rice CM, Mallam EA, Whone AL, Walsh P, Brooks DJ, Kane N, Butler SR, Marks DI, Scolding NJ. Safety and feasibility of autologous bone marrow cellular therapy in relapsing-progressive multiple sclerosis. Clin Pharmacol Ther 2010; 87:679-85. [PMID: 20445531 DOI: 10.1038/clpt.2010.44] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In this phase I study, we assessed the safety and feasibility of intravenous, autologous bone marrow (BM) cell therapy, without immunosuppressive preconditioning, in six patients with clinically definite, relapsing-progressive multiple sclerosis (MS). Assessment of efficacy was a secondary objective and employed clinical disability rating scales, multimodal evoked potential (MMEP) recordings, and magnetic resonance imaging (MRI) scans. Cells were harvested, filtered and infused intravenously in a day-case procedure that was well tolerated by patients and was not associated with any serious adverse events (AEs). Over a period of 12 months after the therapy, clinical disability scores showed either no change (Extended Disability Status Score, EDSS) or improvement (MS impact scale-29, MSIS-29), and MMEPs showed neurophysiological improvement. MRI scans did not show any significant changes over a post-therapy period of 3 months. The lack of serious adverse effects and the suggestion of a beneficial effect in this small sample of patients with progressive disease justify conducting a larger phase II/III study to make a fuller assessment of the efficacy of mobilization of autologous BM in patients with MS.
Collapse
Affiliation(s)
- C M Rice
- Institute of Clinical Neurosciences, University of Bristol, Frenchay Hospital, Bristol, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rice CM, Scolding NJ. Adult human mesenchymal cells proliferate and migrate in response to chemokines expressed in demyelination. Cell Adh Migr 2010; 4:235-40. [PMID: 20234187 DOI: 10.4161/cam.4.2.11404] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Systemic delivery of multipotent mesenchymal stem cells (MSC) may be of benefit in the treatment of neurological diseases, including multiple sclerosis (MS). Certainly, animal studies have demonstrated functional benefits following MSC transplantation, although the mechanisms by which MSCs migrate to lesions and stimulate repair remain unknown. Chemokines stimulate migration in other settings. In this study, we systematically explore the migratory and proliferative responses of human MSCs (hMSC) to chemokines expressed in MS lesions. We demonstrate that these chemokines trigger hMSC migration. In addition, we show that RANTES and IP-10 promote hMSC proliferation.
Collapse
Affiliation(s)
- Claire M Rice
- Institute of Clinical Neurosciences, University of Bristol, Frenchay Hospital, Bristol, UK.
| | | |
Collapse
|