1
|
Wang S, Castro BA, Katz JL, Arrieta V, Najem H, Vazquez-Cervantes GI, Wan H, Olson IE, Hou D, Dapash M, Billingham LK, Chia TY, Wei C, Rashidi A, Platanias LC, McCortney K, Horbinski CM, Stupp R, Zhang P, Ahmed AU, Sonabend AM, Heimberger AB, Lesniak MS, Riviere-Cazaux C, Burns T, Miska J, Fischietti M, Lee-Chang C. B cell-based therapy produces antibodies that inhibit glioblastoma growth. J Clin Invest 2024; 134:e177384. [PMID: 39207859 PMCID: PMC11473152 DOI: 10.1172/jci177384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Glioblastoma (GBM) is a highly aggressive and malignant brain tumor with limited therapeutic options and a poor prognosis. Despite current treatments, the invasive nature of GBM often leads to recurrence. A promising alternative strategy is to harness the potential of the immune system against tumor cells. Our previous data showed that the BVax (B cell-based vaccine) can induce therapeutic responses in preclinical models of GBM. In this study, we aimed to characterize the antigenic reactivity of BVax-derived Abs and evaluate their therapeutic potential. We performed immunoproteomics and functional assays in murine models and samples from patients with GBM. Our investigations revealed that BVax distributed throughout the GBM tumor microenvironment and then differentiated into Ab-producing plasmablasts. Proteomics analyses indicated that the Abs produced by BVax had unique reactivity, predominantly targeting factors associated with cell motility and the extracellular matrix. Crucially, these Abs inhibited critical processes such as GBM cell migration and invasion. These findings provide valuable insights into the therapeutic potential of BVax-derived Abs for patients with GBM, pointing toward a novel direction for GBM immunotherapy.
Collapse
Affiliation(s)
- Si Wang
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Brandyn A. Castro
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Neurological Surgery, University of Chicago Medicine, Chicago, Illinois, USA
| | - Joshua L. Katz
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Victor Arrieta
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Hinda Najem
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Gustavo I. Vazquez-Cervantes
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Hanxiao Wan
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Ian E. Olson
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - David Hou
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mark Dapash
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leah K. Billingham
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Tzu-yi Chia
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Chao Wei
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Aida Rashidi
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
| | - Leonidas C. Platanias
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
- Department of Medicine, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Medicine, Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois, USA
| | - Kathleen McCortney
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Craig M. Horbinski
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Roger Stupp
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Peng Zhang
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Atique U. Ahmed
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Adam M. Sonabend
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Amy B. Heimberger
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | | | - Terry Burns
- Department of Neurological Surgery, Mayo Clinic, Rochester, Minnesotta, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| | - Mariafausta Fischietti
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, Illinois, USA
- Department of Medicine, Division of Hematology and Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Catalina Lee-Chang
- Department of Neurological Surgery, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, USA
- Lou and Jean Malnati Brain Tumor Institute, Chicago, Illinois, USA
| |
Collapse
|
2
|
Seyyedin S, Ezzatabadipour M, Nematollahi-Mahani SN. The Role of Various Factors in Neural Differentiation of Human Umbilical Cord Mesenchymal Stem Cells with a Special Focus on the Physical Stimulants. Curr Stem Cell Res Ther 2024; 19:166-177. [PMID: 36734908 DOI: 10.2174/1574888x18666230124151311] [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: 07/03/2022] [Revised: 10/05/2022] [Accepted: 11/25/2022] [Indexed: 02/04/2023]
Abstract
Human umbilical cord matrix-derived mesenchymal stem cells (hUCMs) are considered as ideal tools for cell therapy procedures and regenerative medicine. The capacity of these cells to differentiate into neural lineage cells make them potentially important in the treatment of various neurodegenerative diseases. An electronic search was performed in Web of Science, PubMed/MEDLINE, Scopus and Google Scholar databases for articles published from January 1990 to March 2022. This review discusses the current knowledge on the effect of various factors, including physical, chemical and biological stimuli which play a key role in the differentiation of hUCMs into neural and glial cells. Moreover, the currently understood molecular mechanisms involved in the neural differentiation of hUCMs under various environmental stimuli are reviewed. Various stimuli, especially physical stimuli and specifically different light sources, have revealed effects on neural differentiation of mesenchymal stem cells, including hUCMs; however, due to the lack of information about the exact mechanisms, there is still a need to find optimal conditions to promote the differentiation capacity of these cells which in turn can lead to significant progress in the clinical application of hUCMs for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Sajad Seyyedin
- Department of Anatomical Sciences, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Massood Ezzatabadipour
- Department of Anatomical Sciences, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Physiology Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Seyed Noureddin Nematollahi-Mahani
- Department of Anatomical Sciences, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
3
|
Luis J, Eastlake K, Lamb WDB, Limb GA, Jayaram H, Khaw PT. Cell-Based Therapies for Glaucoma. Transl Vis Sci Technol 2023; 12:23. [PMID: 37494052 PMCID: PMC10383000 DOI: 10.1167/tvst.12.7.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Glaucomatous optic neuropathy (GON) is the major cause of irreversible visual loss worldwide and can result from a range of disease etiologies. The defining features of GON are retinal ganglion cell (RGC) degeneration and characteristic cupping of the optic nerve head (ONH) due to tissue remodeling, while intraocular pressure remains the only modifiable GON risk factor currently targeted by approved clinical treatment strategies. Efforts to understand the mechanisms that allow species such as the zebrafish to regenerate their retinal cells have greatly increased our understanding of regenerative signaling pathways. However, proper integration within the retina and projection to the brain by the newly regenerated neuronal cells remain major hurdles. Meanwhile, a range of methods for in vitro differentiation have been developed to derive retinal cells from a variety of cell sources, including embryonic and induced pluripotent stem cells. More recently, there has been growing interest in the implantation of glial cells as well as cell-derived products, including neurotrophins, microRNA, and extracellular vesicles, to provide functional support to vulnerable structures such as RGC axons and the ONH. These approaches offer the advantage of not relying upon the replacement of degenerated cells and potentially targeting earlier stages of disease pathogenesis. In order to translate these techniques into clinical practice, appropriate cell sourcing, robust differentiation protocols, and accurate implantation methods are crucial to the success of cell-based therapy in glaucoma. Translational Relevance: Cell-based therapies for glaucoma currently under active development include the induction of endogenous regeneration, implantation of exogenously derived retinal cells, and utilization of cell-derived products to provide functional support.
Collapse
Affiliation(s)
- Joshua Luis
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Karen Eastlake
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - William D. B. Lamb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - G. Astrid Limb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Hari Jayaram
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Peng T. Khaw
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| |
Collapse
|
4
|
Dwivedi S, Choudhary P, Gupta A, Singh S. Therapeutical growth in oligodendroglial fate induction via transdifferentiation of stem cells for neuroregenerative therapy. Biochimie 2023; 211:35-56. [PMID: 36842627 DOI: 10.1016/j.biochi.2023.02.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/20/2022] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
The merits of stem cell therapy and research are undisputed due to their widespread usage in the treatment of neurodegenerative diseases and demyelinating disorders. Cell replacement therapy especially revolves around stem cells and their induction into different cell lineages both adult and progenitor - belonging to each germ layer, prior to transplantation or disease modeling studies. The nervous system is abundant in glial cells and among these are oligodendrocytes capable of myelinating new-born neurons and remyelination of axons with lost or damaged myelin sheath. But demyelinating diseases generate tremendous deficit between myelin loss and recovery. To compensate for this loss, analyze the defects in remyelination mechanisms as well as to trigger full recovery in such patients mesenchymal stem cells (MSCs) have been induced to transdifferentiate into oligodendrocytes. But such experiments are riddled with problems like prolonged, tenuous and complicated protocols that stretch longer than the time taken for the spread of demyelination-associated after-effects. This review delves into such protocols and the combinations of different molecules and factors that have been recruited to derive bona fide oligodendrocytes from in vitro differentiation of embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and MSCs with special focus on MSC-derived oligodendrocytes.
Collapse
Affiliation(s)
- Shrey Dwivedi
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Princy Choudhary
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Ayushi Gupta
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India
| | - Sangeeta Singh
- Department of Applied Sciences, Indian Institute of Information Technology, Allahabad, U.P., India.
| |
Collapse
|
5
|
Combination of Stem Cells with Chinese Herbs for Secondary Depression in Neurodegenerative Diseases Based on Traditional Chinese Medicine Theories. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6847917. [PMID: 35280507 PMCID: PMC8913071 DOI: 10.1155/2022/6847917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 11/15/2021] [Accepted: 01/30/2022] [Indexed: 11/25/2022]
Abstract
Depression is a common secondary symptom in neurodegenerative diseases (NDs) caused by the loss of neurons and glial cells. Recent research focuses on stem cell therapy to replace dead nerve cells, but the low efficiency of stem cell differentiation and short survival time are obstacles limiting the therapy's effectiveness. Clinically, patients with different diseases cannot obtain the same effect by using the same cell therapy. However, traditional Chinese medicine (TCM) often uses syndrome differentiation to determine the treatment plan for NDs. Based on TCM syndrome differentiation and treatment, this article summarizes the advantages of Chinese herbal medicine combined with stem cell therapy, mainly for the effects of various herbs on diseases and stem cells, including prolonging the survival time of stem cells, resisting inflammation, and antidepressant-like effects. In particular, it analyzes the unique pathways of the influence of drugs and acupuncture on different therapies, seeking to clarify the scientific TCM system. This review mainly elaborates on the treatment of secondary depression in TCM and the advantages of a herbal combined stem cell therapy in various methods. We believe it can provide a new clinical concept for secondary depression to obtain good clinical effects and reduce the risks borne by patients.
Collapse
|
6
|
Williams RJ, Tryfonidou MA, Snuggs JW, Le Maitre CL. Cell sources proposed for nucleus pulposus regeneration. JOR Spine 2021; 4:e1175. [PMID: 35005441 PMCID: PMC8717099 DOI: 10.1002/jsp2.1175] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/01/2021] [Accepted: 10/20/2021] [Indexed: 12/13/2022] Open
Abstract
Lower back pain (LBP) occurs in 80% of adults in their lifetime; resulting in LBP being one of the biggest causes of disability worldwide. Chronic LBP has been linked to the degeneration of the intervertebral disc (IVD). The current treatments for chronic back pain only provide alleviation of symptoms through pain relief, tissue removal, or spinal fusion; none of which target regenerating the degenerate IVD. As nucleus pulposus (NP) degeneration is thought to represent a key initiation site of IVD degeneration, cell therapy that specifically targets the restoration of the NP has been reviewed here. A literature search to quantitatively assess all cell types used in NP regeneration was undertaken. With key cell sources: NP cells; annulus fibrosus cells; notochordal cells; chondrocytes; bone marrow mesenchymal stromal cells; adipose-derived stromal cells; and induced pluripotent stem cells extensively analyzed for their regenerative potential of the NP. This review highlights: accessibility; expansion capability in vitro; cell survival in an IVD environment; regenerative potential; and safety for these key potential cell sources. In conclusion, while several potential cell sources have been proposed, iPSC may provide the most promising regenerative potential.
Collapse
Affiliation(s)
- Rebecca J. Williams
- Biomedical Research Centre, BiosciencesSheffield Hallam UniversitySheffieldUK
| | - Marianna A. Tryfonidou
- Department of Clinical Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | | |
Collapse
|
7
|
Human umbilical cord mesenchymal stem cells in type 2 diabetes mellitus: the emerging therapeutic approach. Cell Tissue Res 2021; 385:497-518. [PMID: 34050823 DOI: 10.1007/s00441-021-03461-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 04/11/2021] [Indexed: 12/14/2022]
Abstract
The umbilical cord has been proved to be an easy-access, reliable, and useful source of mesenchymal stem cells (MSC) for clinical applications due to its primitive, immunomodulatory, non-immunogenic, secretory and paracrine, migratory, proliferative, and multipotent properties. This set of characteristics has recently attracted great research interest in the fields of nanotechnology and regenerative medicine and cellular therapy. Accumulating evidence supports a pronounced therapeutic potential of MSC in many different pathologies, from hematology to immunology, wound-healing, tissue regeneration, and oncology. Diabetes mellitus, branded the epidemic of the century, is considered a chronic metabolic disorder, representing a major burden for health system sustainability and an important public health challenge to modern societies. The available treatments for type 2 diabetes mellitus (T2DM) still rely mainly on combinations of oral antidiabetic agents with lifestyle and nutritional adjustments. Despite the continuous development of novel and better hypoglycemic drugs, their efficacy is limited in the installment and progression of silent T2DM complications. T2DM comorbidities and mortality rates still make it a serious, common, costly, and long-term manageable disease. Recently, experimental models, preclinical observations, and clinical studies have provided some insights and preliminary promising results using umbilical cord MSCs to treat and manage diabetes. This review focuses on the latest research and applications of human-derived umbilical cord MSC in the treatment and management of T2DM, exploring and systematizing the key effects of both umbilical cord MSC and its factor-rich secretome accordingly with the major complications associated to T2DM.
Collapse
|
8
|
Ren Z, Qi Y, Sun S, Tao Y, Shi R. Mesenchymal Stem Cell-Derived Exosomes: Hope for Spinal Cord Injury Repair. Stem Cells Dev 2020; 29:1467-1478. [PMID: 33045910 DOI: 10.1089/scd.2020.0133] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a devastating medical condition with profound social and economic impacts. Although research is ongoing, current treatment options are limited and do little to restore functionality. However, recent studies suggest that mesenchymal stem cell-derived exosomes (MSC-exosomes) may hold the key to exciting new treatment options for SCI patients. MSCs are self-renewing multipotent stem cells with multi-directional differentiation and can secrete a large number of exosomes (vesicles secreted into the extracellular environment through endocytosis, called MSC-exosomes). These MSC-exosomes play a critical role in repairing SCI through promoting angiogenesis and axonal growth, regulating inflammation and the immune response, inhibiting apoptosis, and maintaining the integrity of the blood-spinal cord barrier. Furthermore, they can be utilized to transport genetic material or drugs to target cells, and their relatively small size makes them able to permeate the blood-brain barrier. In this review, we summarize recent advances in MSC-exosome themed SCI treatments and cell-free therapies to better understand this newly emerging methodology.
Collapse
Affiliation(s)
- Zhihua Ren
- Department of Basic Medical Sciences, College of Veterinary Medicine, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Yaan, China
| | - Yao Qi
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Yaan, China
| | - Siyuan Sun
- Department of Basic Medical Sciences, College of Veterinary Medicine, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA.,Department of Orthopedics, National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuanyuan Tao
- Department of Basic Medical Sciences, College of Veterinary Medicine, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA.,Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Yaan, China
| | - Riyi Shi
- Department of Basic Medical Sciences, College of Veterinary Medicine, Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, USA
| |
Collapse
|
9
|
Khan AA, Huat TJ, Al Mutery A, El-Serafi AT, Kacem HH, Abdallah SH, Reza MF, Abdullah JM, Jaafar H. Significant transcriptomic changes are associated with differentiation of bone marrow-derived mesenchymal stem cells into neural progenitor-like cells in the presence of bFGF and EGF. Cell Biosci 2020; 10:126. [PMID: 33133516 PMCID: PMC7594431 DOI: 10.1186/s13578-020-00487-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Mesenchymal stem cells (MSCs) isolated from bone marrow have different developmental origins, including neural crest. MSCs can differentiate into neural progenitor-like cells (NPCs) under the influence of bFGF and EGF. NPCs can terminally differentiate into neurons that express beta-III-tubulin and elicit action potential. The main aim of the study was to identify key genetic markers involved in differentiation of MSCs into NPCs through transcriptomic analysis. METHOD Total RNA was isolated from MSCs and MSCs-derived NPCs followed by cDNA library construction for transcriptomic analysis. Sample libraries that passed the quality and quantity assessments were subjected to high throughput mRNA sequencing using NextSeq®500. Differential gene expression analysis was performed using the DESeq2 R package with MSC samples being a reference group. The expression of eight differentially regulated genes was counter validated using real-time PCR. RESULTS In total, of the 3,252 differentially regulated genes between MSCs and NPCs with two or more folds, 1,771 were upregulated genes, whereas 1,481 were downregulated in NPCs. Amongst these differential genes, 104 transcription factors were upregulated, and 45 were downregulated in NPCs. Neurogenesis related genes were upregulated in NPCs and the main non-redundant gene ontology (GO) terms enriched in NPCs were the autonomic nervous system, cell surface receptor signalling pathways), extracellular structure organisation, and programmed cell death. The main non-redundant GO terms enriched in MSCs included cytoskeleton organisation cytoskeleton structural constituent, mitotic cell cycle), and the mitotic cell cycle process Gene set enrichment analysis also confirmed cell cycle regulated pathways as well as Biocarta integrin pathway were upregulated in MSCs. Transcription factors enrichment analysis by ChEA3 revealed Foxs1 and HEYL, amongst the top five transcription factors, inhibits and enhances, respectively, the NPCs differentiation of MSCs. CONCLUSIONS The vast differences in the transcriptomic profiles between NPCs and MSCs revealed a set of markers that can identify the differentiation stage of NPCs as well as provide new targets to enhance MSCs differentiation into NPCs.
Collapse
Affiliation(s)
- Amir Ali Khan
- Department of Applied Biology, College of Sciences, University of Sharjah, P.O. Box 27272, Emirate of Sharjah, United Arab Emirates
- Research Institute of Science and Engineering, University of Sharjah, P.O. Box 27272, Emirate of Sharjah, United Arab Emirates
| | - Tee Jong Huat
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543 Singapore
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Jalan Raja Perempuan Zainab II, 16150, Kubang Kerian, Kelantan Malaysia
| | - Abdullah Al Mutery
- Department of Applied Biology, College of Sciences, University of Sharjah, P.O. Box 27272, Emirate of Sharjah, United Arab Emirates
| | - Ahmed Taher El-Serafi
- Department of Biomedical and Clinical Sciences (BKV), Linköping University, P.O. Box 581 83, Linköping, Sweden
| | - Hassen Hadj Kacem
- Department of Applied Biology, College of Sciences, University of Sharjah, P.O. Box 27272, Emirate of Sharjah, United Arab Emirates
- Research Institute of Science and Engineering, University of Sharjah, P.O. Box 27272, Emirate of Sharjah, United Arab Emirates
| | - Sallam Hasan Abdallah
- Research Institute of Science and Engineering, University of Sharjah, P.O. Box 27272, Emirate of Sharjah, United Arab Emirates
| | - Muhammed Faruque Reza
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Jalan Raja Perempuan Zainab II, 16150, Kubang Kerian, Kelantan Malaysia
| | - Jafri Malin Abdullah
- Department of Neuroscience, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Jalan Raja Perempuan Zainab II, 16150, Kubang Kerian, Kelantan Malaysia
- Brain and Behavior Cluster, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Jalan Raja Perempuan Zainab II, 16150, Kubang Kerian, Kelantan Malaysia
| | - Hasnan Jaafar
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia Health Campus, Jalan Raja Perempuan Zainab II, 16150, Kubang Kerian, Kelantan Malaysia
| |
Collapse
|
10
|
Effects and Mechanisms of Acupuncture Combined with Mesenchymal Stem Cell Transplantation on Neural Recovery after Spinal Cord Injury: Progress and Prospects. Neural Plast 2020; 2020:8890655. [PMID: 33061954 PMCID: PMC7533022 DOI: 10.1155/2020/8890655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/29/2020] [Accepted: 09/05/2020] [Indexed: 02/07/2023] Open
Abstract
Spinal cord injury (SCI) is a structural event with devastating consequences worldwide. Due to the limited intrinsic regenerative capacity of the spinal cord in adults, the neural restoration after SCI is difficult. Acupuncture is effective for SCI-induced neurologic deficits, and the potential mechanisms responsible for its effects involve neural protection by the inhibition of inflammation, oxidation, and apoptosis. Moreover, acupuncture promotes neural regeneration and axon sprouting by activating multiple cellular signal transduction pathways, such as the Wnt, Notch, and Rho/Rho kinase (ROCK) pathways. Several studies have demonstrated that the efficacy of combining acupuncture with mesenchymal stem cells (MSCs) transplantation is superior to either procedure alone. The advantage of the combined treatment is dependent on the ability of acupuncture to enhance the survival of MSCs, promote their differentiation into neurons, and facilitate targeted migration of MSCs to the spinal cord. Additionally, the differentiation of MSCs into neurons overcomes the problem of the shortage of endogenous neural stem cells (NSCs) in the acupuncture-treated SCI patients. Therefore, the combination of acupuncture and MSCs transplantation could become a novel and effective strategy for the treatment of SCI. Such a possibility needs to be verified by basic and clinical research.
Collapse
|
11
|
Domingues C, Geraldo AM, Anjo SI, Matos A, Almeida C, Caramelo I, Lopes-da-Silva JA, Paiva A, Carvalho J, Pires das Neves R, Manadas B, Grãos M. Cofilin-1 Is a Mechanosensitive Regulator of Transcription. Front Cell Dev Biol 2020; 8:678. [PMID: 32903827 PMCID: PMC7438942 DOI: 10.3389/fcell.2020.00678] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
The mechanical properties of the extracellular environment are interrogated by cells and integrated through mechanotransduction. Many cellular processes depend on actomyosin-dependent contractility, which is influenced by the microenvironment’s stiffness. Here, we explored the influence of substrate stiffness on the proteome of proliferating undifferentiated human umbilical cord-matrix mesenchymal stem/stromal cells. The relative abundance of several proteins changed significantly by expanding cells on soft (∼3 kPa) or stiff substrates (GPa). Many such proteins are associated with the regulation of the actin cytoskeleton, a major player of mechanotransduction and cell physiology in response to mechanical cues. Specifically, Cofilin-1 levels were elevated in cells cultured on soft comparing with stiff substrates. Furthermore, Cofilin-1 was de-phosphorylated (active) and present in the nuclei of cells kept on soft substrates, in contrast with phosphorylated (inactive) and widespread distribution in cells on stiff. Soft substrates promoted Cofilin-1-dependent increased RNA transcription and faster RNA polymerase II-mediated transcription elongation. Cofilin-1 is part of a novel mechanism linking mechanotransduction and transcription.
Collapse
Affiliation(s)
- Catarina Domingues
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - A Margarida Geraldo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Sandra Isabel Anjo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - André Matos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Cláudio Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Polytechnic Institute of Coimbra, Coimbra College of Agriculture, Coimbra, Portugal
| | - Inês Caramelo
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | | | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Instituto Politécnico de Coimbra, ESTESC-Coimbra Health School, Ciências Biomédicas Laboratoriais, Coimbra, Portugal
| | - João Carvalho
- Centro de Física da Universidade de Coimbra (CFisUC), Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Ricardo Pires das Neves
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - Bruno Manadas
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Mário Grãos
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.,Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal.,Biocant, Technology Transfer Association, Cantanhede, Portugal
| |
Collapse
|
12
|
Mehdipour A, Ebrahimi A, Shiri-Shahsavar MR, Soleimani-Rad J, Roshangar L, Samiei M, Ebrahimi-Kalan A. The potentials of umbilical cord-derived mesenchymal stem cells in the treatment of multiple sclerosis. Rev Neurosci 2020; 30:857-868. [PMID: 31026226 DOI: 10.1515/revneuro-2018-0057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 02/15/2019] [Indexed: 12/12/2022]
Abstract
Stem cell therapy has indicated a promising treatment capacity for tissue regeneration. Multiple sclerosis is an autoimmune-based chronic disease, in which the myelin sheath of the central nervous system is destructed. Scientists have not discovered any cure for multiple sclerosis, and most of the treatments are rather palliative. The pursuit of a versatile treatment option, therefore, seems essential. The immunoregulatory and non-chronic rejection characteristics of mesenchymal stem cells, as well as their homing properties, recommend them as a prospective treatment option for multiple sclerosis. Different sources of mesenchymal stem cells have distinct characteristics and functional properties; in this regard, choosing the most suitable cell therapy approach seems to be challenging. In this review, we will discuss umbilical cord/blood-derived mesenchymal stem cells, their identified exclusive properties compared to another adult mesenchymal stem cells, and the expectations of their potential roles in the treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Ahmad Mehdipour
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ayyub Ebrahimi
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Haliç University, Istanbul, Turkey
| | | | - Jafar Soleimani-Rad
- Department of Anatomical Sciences, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Roshangar
- Department of Anatomical Sciences, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Samiei
- Endodontics Department of Dental Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi-Kalan
- Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Radiology, School of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran,
| |
Collapse
|
13
|
Slanzi A, Iannoto G, Rossi B, Zenaro E, Constantin G. In vitro Models of Neurodegenerative Diseases. Front Cell Dev Biol 2020; 8:328. [PMID: 32528949 PMCID: PMC7247860 DOI: 10.3389/fcell.2020.00328] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are progressive degenerative conditions characterized by the functional deterioration and ultimate loss of neurons. These incurable and debilitating diseases affect millions of people worldwide, and therefore represent a major global health challenge with severe implications for individuals and society. Recently, several neuroprotective drugs have failed in human clinical trials despite promising pre-clinical data, suggesting that conventional cell cultures and animal models cannot precisely replicate human pathophysiology. To bridge the gap between animal and human studies, three-dimensional cell culture models have been developed from human or animal cells, allowing the effects of new therapies to be predicted more accurately by closely replicating some aspects of the brain environment, mimicking neuronal and glial cell interactions, and incorporating the effects of blood flow. In this review, we discuss the relative merits of different cerebral models, from traditional cell cultures to the latest high-throughput three-dimensional systems. We discuss their advantages and disadvantages as well as their potential to investigate the complex mechanisms of human neurodegenerative diseases. We focus on in vitro models of the most frequent age-related neurodegenerative disorders, such as Parkinson’s disease, Alzheimer’s disease and prion disease, and on multiple sclerosis, a chronic inflammatory neurodegenerative disease affecting young adults.
Collapse
Affiliation(s)
- Anna Slanzi
- Department of Medicine, University of Verona, Verona, Italy
| | - Giulia Iannoto
- Department of Medicine, University of Verona, Verona, Italy
| | - Barbara Rossi
- Department of Medicine, University of Verona, Verona, Italy
| | - Elena Zenaro
- Department of Medicine, University of Verona, Verona, Italy
| | - Gabriela Constantin
- Department of Medicine, University of Verona, Verona, Italy.,Center for Biomedical Computing (CBMC), University of Verona, Verona, Italy
| |
Collapse
|
14
|
Borhani-Haghighi M, Mohamadi Y. Intranasal administration of conditioned medium derived from mesenchymal stem cells-differentiated oligodendrocytes ameliorates experimental autoimmune encephalomyelitis. J Chem Neuroanat 2020; 106:101792. [PMID: 32353514 DOI: 10.1016/j.jchemneu.2020.101792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/05/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022]
Abstract
In multiple sclerosis, myelin sheaths around the axons are degenerated due to uncontrolled inflammation in the central nervous system. Oligodendrocytes (OLs) are myelin-forming cells that secrete trophic factors necessary for myelin protection. Beneficial features of conditioned medium (CM) derived from different stem cells are nowadays under investigation in treating neurodegenerative diseases. Here, we used the differentiation capacity of Wharton's jelly mesenchymal stem cells (WJMSCs) to obtain OLs. Then, the study aimed to evaluate the status of inflammation and myelination in male experimental autoimmune encephalomyelitis (EAE) mice after intranasal administration of CM derived from OLs (OL-CM). Inflammation was studied by evaluating gliosis, inflammatory cell infiltration and expression of inflammation indicators including NLRP3 inflammasome, interleukin-1β, interleukin-18, glial fibrillary acidic protein, and ionized calcium binding adaptor molecule 1. Remyelination was studied by luxol fast blue staining and evaluating the expression of myelin indicators including myelin basic protein and oligodendrocyte transcription factor. In addition, we followed the trend of body weight and functional recovery during the 28-day study. ELISA assay revealed that OL-CM contained brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, and ciliary neurotrophic factor. Data showed that OL-CM moderated inflammation, augmented remyelination, and gained normal body weight. Notably, these anti-inflammatory and regenerative effects of OL-CM improved neurological functions in EAE mice. In conclusion, the current study offered a new choice for treating multiple sclerosis using noninvasive intranasal administration of CM harvested from easily achievable WJMSCs-differentiated OLs.
Collapse
Affiliation(s)
- Maryam Borhani-Haghighi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Mohamadi
- Department of Anatomy, School of Medicine, Ilam University of Medical Sciences, Ilam, Iran.
| |
Collapse
|
15
|
Kruminis-Kaszkiel E, Osowski A, Bejer-Oleńska E, Dziekoński M, Wojtkiewicz J. Differentiation of Human Mesenchymal Stem Cells from Wharton's Jelly Towards Neural Stem Cells Using A Feasible and Repeatable Protocol. Cells 2020; 9:cells9030739. [PMID: 32192154 PMCID: PMC7140706 DOI: 10.3390/cells9030739] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 03/14/2020] [Accepted: 03/15/2020] [Indexed: 12/15/2022] Open
Abstract
The transplantation of neural stem cells (NSCs) capable of regenerating to the cells of the central nervous system (CNS) is a promising strategy in the treatment of CNS diseases and injury. As previous studies have highlighted mesenchymal stem cells (MSCs) as a source of NSCs, this study aimed to develop a feasible, efficient, and reproducible method for the neural induction of MSCs isolated from Wharton's jelly (hWJ-MSCs). We induced neural differentiation in a monolayer culture using epidermal growth factor, basic fibroblast growth factor, N2, and B27 supplements. This resulted in a homogenous population of proliferating cells that expressed certain neural markers at both the protein and mRNA levels. Flow cytometry and immunocytochemistry confirmed the expression of neural markers: nestin, sex-determining region Y (SRY) box 1 and 2 (SOX1 and SOX2), microtubule-associated protein 2 (MAP2), and glial fibrillary acidic protein (GFAP). The qRT-PCR analysis revealed significantly enhanced expression of nestin and MAP2 in differentiated cells. This study confirms that it is possible to generate NSCs-like cells from hWJ-MSCs in a 2D culture using a practical method. However, the therapeutic effectiveness of such differentiated cells should be extended to confirm the terminal differentiation ability and electrophysiological properties of neurons derived from them.
Collapse
Affiliation(s)
- Ewa Kruminis-Kaszkiel
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
- Correspondence:
| | - Adam Osowski
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
| | - Ewa Bejer-Oleńska
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
| | - Mariusz Dziekoński
- Department of Animal Anatomy and Physiology, Faculty of Biology and Biotechnology, University of Warmia and Mazury, 10-719 Olsztyn, Poland;
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (A.O.); (E.B.-O.); (J.W.)
| |
Collapse
|
16
|
Hedrick BP, Dickson BV, Dumont ER, Pierce SE. The evolutionary diversity of locomotor innovation in rodents is not linked to proximal limb morphology. Sci Rep 2020; 10:717. [PMID: 31959908 PMCID: PMC6970985 DOI: 10.1038/s41598-019-57144-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 12/11/2019] [Indexed: 01/08/2023] Open
Abstract
Rodents are the most species-rich order within Mammalia and have evolved disparate morphologies to accommodate numerous locomotor niches, providing an excellent opportunity to understand how locomotor innovation can drive speciation. To evaluate the connection between the evolutionary success of rodents and the diversity of rodent locomotor ecologies, we used a large dataset of proximal limb CT scans from across Myomorpha and Geomyoidea to examine internal and external limb shape. Only fossorial rodents displayed a major reworking of their proximal limbs in either internal or external morphology, with other locomotor modes plotting within a generalist morphospace. Fossorial rodents were also the only locomotor mode to consistently show increased rates of humerus/femur morphological evolution. We propose that these rodent clades were successful at spreading into ecological niches due to high behavioral plasticity and small body sizes, allowing them to modify their locomotor mode without requiring major changes to their proximal limb morphology.
Collapse
Affiliation(s)
- Brandon P Hedrick
- Department of Cell Biology and Anatomy, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA.
- Museum of Comparative Zoology and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA.
- Department of Earth Sciences, University of Oxford, Oxford, UK.
| | - Blake V Dickson
- Museum of Comparative Zoology and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Elizabeth R Dumont
- School of Natural Sciences, University of California-Merced, Merced, CA, 95343, USA
| | - Stephanie E Pierce
- Museum of Comparative Zoology and Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
17
|
Ma J, Li J, Wang Y, Chen W, Zheng P, Chen Y, Sun Z, Liu J, Zhou Y, Wang J, Liu S, Han X. WSZG inhibits BMSC-induced EMT and bone metastasis in breast cancer by regulating TGF-β1/Smads signaling. Biomed Pharmacother 2019; 121:109617. [PMID: 31810139 DOI: 10.1016/j.biopha.2019.109617] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/18/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023] Open
Abstract
Bone metastasis of breast cancer causes severe skeletal-related events and poor prognosis. Wensheng Zhuanggu Formula (WSZG), a traditional Chinese prescription, is used to adjunctively treat breast cancer bone metastases in clinical practice. This study was undertaken to investigate the antibone-metastatic activities and mechanisms of WSZG extract by evaluating the effect of this formula on the cross-talk between bone marrow-derived mesenchymal stem cells (BMSCs) and breast cancer cells in triggering epithelial-mesenchymal transition (EMT) in vivo and in vitro. The results demonstrated that BMSCs might enhance the invasive and metastatic potentials of breast cancer cells as a consequence of EMT induction through direct cell-to-cell contact. WSZG treatment remarkably suppressed motility, invasion, EMT-related gene, and protein markers in BMSC-conditioned breast cancer cells and ameliorated bone metastases and damages in nude mice following co-injection of BMSCs and MDA-MB-231BO breast cancer cells. Further investigation showed that the transforming growth factor-β1 (TGF-β1)/Smads pathway was an important mechanism enabling BMSCs to induce EMT occurrence of breast cancer cells. WSZG treatment reversed BMSC-induced EMT by downregulating TGF-β1/Smads signaling. Thus, WSZG extracts may be regarded as a potential antibone-metastatic agent for breast cancer therapy.
Collapse
Affiliation(s)
- Jiao Ma
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jiajia Li
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ying Wang
- Shanghai University of Traditional Chinese Medicine, School of Chinese Materia Medica, Shanghai 201203, China
| | - Weiling Chen
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Peiyong Zheng
- Institute of Digestive Diseases, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yueqiang Chen
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Zhenping Sun
- Department of Breast Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jin Liu
- Department of Breast Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yin Zhou
- Department of Breast Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jianyi Wang
- Department of Liver Disease, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Sheng Liu
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Xianghui Han
- Institute of Chinese Traditional Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
18
|
Sanabria-Castro A, Flores-Díaz M, Alape-Girón A. Biological models in multiple sclerosis. J Neurosci Res 2019; 98:491-508. [PMID: 31571267 DOI: 10.1002/jnr.24528] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 09/05/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022]
Abstract
Considering the etiology of multiple sclerosis (MS) is still unknown, experimental models resembling specific aspects of this immune-mediated demyelinating human disease have been developed to increase the understanding of processes related to pathogenesis, disease evolution, evaluation of therapeutic interventions, and demyelination and remyelination mechanisms. Based on the nature of the investigation, biological models may include in vitro, in vivo, and ex vivo assessments. Even though these approaches have disclosed valuable information, every disease animal model has limitations and can only replicate specific features of MS. In vitro and ex vivo models generally do not reflect what occurs in the organism, and in vivo animal models are more likely used; nevertheless, they are able to reproduce only certain stages of the disease. In vivo MS disease animal models in mammals include: experimental autoimmune encephalomyelitis, viral encephalomyelitis, and induced demyelination. This review examines and describes the most common biological disease animal models for the study of MS, their specific characteristics and limitations.
Collapse
Affiliation(s)
- Alfredo Sanabria-Castro
- Research Unit, San Juan de Dios Hospital CCSS, San José, Costa Rica.,School of Pharmacy, University of Costa Rica, San José, Costa Rica
| | | | | |
Collapse
|
19
|
Soft culture substrates favor stem-like cellular phenotype and facilitate reprogramming of human mesenchymal stem/stromal cells (hMSCs) through mechanotransduction. Sci Rep 2019; 9:9086. [PMID: 31235788 PMCID: PMC6591285 DOI: 10.1038/s41598-019-45352-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 06/04/2019] [Indexed: 01/26/2023] Open
Abstract
Biophysical cues influence many aspects of cell behavior. Stiffness of the extracellular matrix is probed by cells and transduced into biochemical signals through mechanotransduction protein networks, strongly influencing stem cell behavior. Cellular stemness is intimately related with mechanical properties of the cell, like intracellular contractility and stiffness, which in turn are influenced by the microenvironment. Pluripotency is associated with soft and low-contractility cells. Hence, we postulated that soft cell culture substrates, presumably inducing low cellular contractility and stiffness, increase the reprogramming efficiency of mesenchymal stem/stromal cells (MSCs) into induced pluripotent stem cells (iPSCs). We demonstrate that soft substrates (1.5 or 15 kPa polydimethylsiloxane – PDMS) caused modulation of several cellular features of MSCs into a phenotype closer to pluripotent stem cells (PSCs). MSCs cultured on soft substrates presented more relaxed nuclei, lower maturation of focal adhesions and F-actin assembling, more euchromatic and less heterochromatic nuclear DNA regions, and increased expression of pluripotency-related genes. These changes correlate with the reprogramming of MSCs, with a positive impact on the kinetics, robustness of colony formation and reprogramming efficiency. Additionally, substrate stiffness influences several phenotypic features of iPS cells and colonies, and data indicates that soft substrates favor full iPSC reprogramming.
Collapse
|
20
|
Liu Y, Given KS, Dickson EL, Owens GP, Macklin WB, Bennett JL. Concentration-dependent effects of CSF1R inhibitors on oligodendrocyte progenitor cells ex vivo and in vivo. Exp Neurol 2019; 318:32-41. [PMID: 31029597 DOI: 10.1016/j.expneurol.2019.04.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/21/2019] [Accepted: 04/22/2019] [Indexed: 01/18/2023]
Abstract
Microglia are the principal resident immune cells in the central nervous system (CNS) and play important roles in CNS development, maintenance and repair. The survival and development of microglia depends on colony-stimulating factor 1 receptor (CSF1R), a member of the platelet-derived growth factor receptor (PDGFR) family of tyrosine kinases. Recently pharmacological CSF1R inhibition has been used to investigate the effects of microglial depletion in numerous animal models of CNS disease. However, the effects of CSF1R inhibitors on other cell types in the CNS remains incompletely characterized. In this report, we compared the effect of two commonly used CSF1R inhibitors, PLX5622 and PLX3397, on microglia and oligodendrocyte progenitor cell (OPC) numbers. In ex vivo cerebellar slices and adult mouse brain, both PLX compounds caused robust microglia loss; the kinetics of microglial depletion was more rapid with PLX5622. While high-doses of PLX5622 and PLX3397 reduced OPC number in primary cultures in vitro and ex vivo, low-doses of PLX5622 did not affect the number of OPCs or mature oligodendroglia in culture or in vivo. In adult mice, treatment with PLX5622 had no effect on OPC numbers for 7 days; however, a mild reduction was observed after 21 days in some CNS regions. In contrast, PLX3397 caused significant OPC loss after 7 days of treatment, despite only modest microglia depletion. Neither PLX compound had a remarkable effect on mature oligodendrocytes or myelin protein expression following long-term oral administration. Our results show that CSF1R inhibition with PLX5622 can selectively deplete microglia ex vivo and in vivo without affecting OPC number, demonstrating that microglia are not essential for OPC viability in ex vivo slice cultures or adult CNS tissues.
Collapse
Affiliation(s)
- Yiting Liu
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA.
| | - Katherine S Given
- Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Erin L Dickson
- Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Gregory P Owens
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Wendy B Macklin
- Department of Cell & Developmental Biology, University of Colorado School of Medicine, Aurora, CO, USA; Program in Neuroscience, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jeffrey L Bennett
- Department of Neurology, University of Colorado School of Medicine, Aurora, CO, USA; Department of Ophthalmology, University of Colorado School of Medicine, Aurora, CO, USA; Program in Neuroscience, University of Colorado School of Medicine, Aurora, CO, USA.
| |
Collapse
|
21
|
Urrutia DN, Caviedes P, Mardones R, Minguell JJ, Vega-Letter AM, Jofre CM. Comparative study of the neural differentiation capacity of mesenchymal stromal cells from different tissue sources: An approach for their use in neural regeneration therapies. PLoS One 2019; 14:e0213032. [PMID: 30856179 PMCID: PMC6437714 DOI: 10.1371/journal.pone.0213032] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 02/13/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can trans/differentiate to neural precursors and/or
mature neurons and promote neuroprotection and neurogenesis. The above could
greatly benefit neurodegenerative disorders as well as in the treatment of
post-traumatic and hereditary diseases of the central nervous system (CNS). In
order to attain an ideal source of adult MSCs for the treatment of CNS diseases,
adipose tissue, bone marrow, skin and umbilical cord derived MSCs were isolated
and studied to explore differences with regard to neural differentiation
capacity. In this study, we demonstrated that MSCs from several tissues can
differentiate into neuron-like cells and differentially express progenitors and
mature neural markers. Adipose tissue MSCs exhibited significantly higher
expression of neural markers and had a faster proliferation rate. Our results
suggest that adipose tissue MSCs are the best candidates for the use in
neurological diseases.
Collapse
Affiliation(s)
| | - Pablo Caviedes
- Program of Molecular & Clinical Pharmacology, ICBM, Faculty of
Medicine, Universidad de Chile, Santiago, Chile
- Centro de Biotecnología y Bioingeniería (CeBiB), Departamento de
Ingeniería Química, Biotecnología y Materiales, Facultad de Ciencias Físicas y
Matemáticas, Universidad de Chile, Santiago, Chile
| | - Rodrigo Mardones
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
- Orthopedic Department, Clinica Las Condes, Santiago,
Chile
| | - José J. Minguell
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
| | - Ana Maria Vega-Letter
- Program of Traslational Immunology ICIM, Faculty of Medicine, Clinica
Alemana Universidad del Desarrollo, Santiago, Chile
| | - Claudio M. Jofre
- Regenerative Cell Therapy Center, Clinica Las Condes, Santiago,
Chile
- * E-mail:
| |
Collapse
|
22
|
Rao R, Bora R. Timing of Umbilical Cord Clamping and Infant Brain Development. J Pediatr 2018; 203:8-10. [PMID: 30193752 DOI: 10.1016/j.jpeds.2018.07.080] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/24/2018] [Indexed: 12/28/2022]
Affiliation(s)
- Raghavendra Rao
- Division of Neonatology Department of Pediatrics University of Minnesota Minneapolis, Minnesota.
| | - Reeta Bora
- Neonatal Unit Department of Pediatrics Assam Medical College Dibrugarh, Assam, India
| |
Collapse
|
23
|
Abiko M, Mitsuhara T, Okazaki T, Imura T, Nakagawa K, Otsuka T, Oshita J, Takeda M, Kawahara Y, Yuge L, Kurisu K. Rat Cranial Bone-Derived Mesenchymal Stem Cell Transplantation Promotes Functional Recovery in Ischemic Stroke Model Rats. Stem Cells Dev 2018; 27:1053-1061. [PMID: 29786481 DOI: 10.1089/scd.2018.0022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The functional disorders caused by central nervous system (CNS) diseases, such as ischemic stroke, are clinically incurable and current treatments have limited effects. Previous studies suggested that cell-based therapy using mesenchymal stem cells (MSCs) exerts therapeutic effects for ischemic stroke. In addition, the characteristics of MSCs may depend on their sources. Among the derived tissues of MSCs, we have focused on cranial bones originating from the neural crest. We previously demonstrated that the neurogenic potential of human cranial bone-derived MSCs (cMSCs) was higher than that of human iliac bone-derived MSCs. Therefore, we presumed that cMSCs have a higher therapeutic potential for CNS diseases. However, the therapeutic effects of cMSCs have not yet been elucidated in detail. In the present study, we aimed to demonstrate the therapeutic effects of transplantation with rat cranial bone-derived MSCs (rcMSCs) in ischemic stroke model rats. The mRNA expression of brain-derived neurotrophic factor and nerve growth factor was significantly stronger in rcMSCs than in rat bone marrow-derived MSCs (rbMSCs). Ischemic stroke model rats in the rcMSC transplantation group showed better functional recovery than those in the no transplantation and rbMSC transplantation groups. Furthermore, in the in vitro study, the conditioned medium of rcMSCs significantly suppressed the death of neuroblastoma × glioma hybrid cells (NG108-15) exposed to oxidative and inflammatory stresses. These results suggest that cMSCs have potential as a candidate cell-based therapy for CNS diseases.
Collapse
Affiliation(s)
- Masaru Abiko
- 1 Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Takafumi Mitsuhara
- 1 Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Takahito Okazaki
- 1 Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Takeshi Imura
- 2 Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Kei Nakagawa
- 2 Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Takashi Otsuka
- 2 Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Jumpei Oshita
- 1 Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Masaaki Takeda
- 1 Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| | - Yumi Kawahara
- 3 Space Bio-Laboratories Co., Ltd. , Hiroshima, Japan
| | - Louis Yuge
- 2 Division of Bio-Environmental Adaptation Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan .,3 Space Bio-Laboratories Co., Ltd. , Hiroshima, Japan
| | - Kaoru Kurisu
- 1 Department of Neurosurgery, Graduate School of Biomedical and Health Sciences, Hiroshima University , Hiroshima, Japan
| |
Collapse
|
24
|
Mukai T, Tojo A, Nagamura-Inoue T. Umbilical Cord-Derived Mesenchymal Stromal Cells Contribute to Neuroprotection in Neonatal Cortical Neurons Damaged by Oxygen-Glucose Deprivation. Front Neurol 2018; 9:466. [PMID: 29963009 PMCID: PMC6013549 DOI: 10.3389/fneur.2018.00466] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 05/31/2018] [Indexed: 12/11/2022] Open
Abstract
Several studies have reported that human umbilical cord-derived mesenchymal stromal cells (UC-MSCs) restore neurological damage in vivo through their secretion of paracrine factors. We previously found that UC-MSCs attenuate brain injury by secreting neurotrophic factors, such as brain-derived neurotrophic factor (BDNF) and hepatocyte growth factor (HGF). However, how these factors contribute to neuroprotection remains unknown. In this study, we aimed to investigate to what extent UC-MSC-derived HGF and BDNF contribute to neuroprotection using a Transwell co-culture system of neonatal cortical neurons damaged by oxygen-glucose deprivation. The influence of HGF and BDNF were determined by investigating neurons in both the presence and absence of UC-MSCs as these cells consistently secrete both factors and can be blocked by neutralizing antibodies. In the co-culture, UC-MSCs significantly improved neuronal injury, as indicated by an increase in immature neuron number, neurite outgrowth, and cell proliferation. Co-culture of damaged neurons with UC-MSCs also exhibited a reduction in the number of neurons displaying signs of apoptosis/necrosis. The neuroprotective actions of UC-MSCs were partially reverted by neutralizing antibodies. Together, our findings reveal that UC-MSC-secreted HGF and BDNF have neuroprotective effects on damaged neurons. Further studies should address the existence of other potential neurotrophic paracrine factors.
Collapse
Affiliation(s)
- Takeo Mukai
- Division of Molecular of Therapy, Center for Advanced Medical Research, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Cell Processing and Transfusion, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Arinobu Tojo
- Division of Molecular of Therapy, Center for Advanced Medical Research, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Department of Cell Processing and Transfusion, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
25
|
Tian KW, Zhang YY, Jiang H, Han S. Intravenous C16 and angiopoietin-1 improve the efficacy of placenta-derived mesenchymal stem cell therapy for EAE. Sci Rep 2018; 8:4649. [PMID: 29545630 PMCID: PMC5854598 DOI: 10.1038/s41598-018-22867-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 02/08/2018] [Indexed: 02/07/2023] Open
Abstract
The placenta has emerged as an attractive source of mesenchymal stem cells (MSCs) because of the absence of ethical issues, non-invasive access, and abundant yield. However, inflammatory cell invasion into grafts negatively impacts the survival and efficacy of transplanted cells. Previous studies have shown that synthetic C16 peptide can competitively block the transmigration of leukocytes into the central nerve system, while angiopoietin-1 (Ang-1) can inhibit inflammation-induced blood vessel leakage and inflammatory cell infiltration in rats with experimental allergic encephalomyelitis (EAE). In this study, we investigated the effects of intravenous administration of C16 and Ang-1 on the efficacy of placenta-derived MSC (PMSC) transplantation in a rat model of EAE. We found that, compared with PMSCs alone, treatment with PMSCs along with intravenously administered C16 and Ang-1 was more effective at ameliorating demyelination/neuronal loss and neurological dysfunction, reducing inflammatory cell infiltration, perivascular edema, and reactive astrogliosis (p < 0.05). Mechanistic studies revealed that intravenous C16 and Ang-1 increased PMSC engraftment in the central nervous system and promoted expression of the neurotropic proteins brain-derived neurotrophic factor, growth-associated protein 43, and p75 neurotrophin receptor as well as the neuronal-glial lineage markers neurofilament protein 200 and myelin basic protein in the engrafted PMSCs.
Collapse
Affiliation(s)
- Ke-Wei Tian
- Institute of Anatomy and Cell Biology, Medical College, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, China
| | - Yuan-Yuan Zhang
- Institute of Anatomy and Cell Biology, Medical College, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, China
| | - Hong Jiang
- Department of Electrophysiology, SirRunRunShaw Hospital, Medical College, Zhejiang University, 310016, Hangzhou, Zhejiang Province, China
| | - Shu Han
- Institute of Anatomy and Cell Biology, Medical College, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, China.
| |
Collapse
|
26
|
Ghasemi N. Transdifferentiation of human adipose-derived mesenchymal stem cells into oligodendrocyte progenitor cells. IRANIAN JOURNAL OF NEUROLOGY 2018; 17:24-30. [PMID: 30186556 PMCID: PMC6121205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Background: Stem cell-based therapy is a new method for the treatment of neurodegenerative diseases such as multiple sclerosis (MS). Human adipose-derived stem cells (hADSCs) are a kind of adult stem cells which have a higher frequency in the fat tissue and have the ability to differentiate into other cell types outside their lineage. Due to some serious adverse events of cell-based therapy such as tumorigenic potential, the aim of this study was to evaluate of hADSCs differentiation into oligodendrocytes as a valuable way for future cell transplantation. Methods: hADSC were isolated from lipoaspirate samples of human abdominal fat. After hADSC characterization via flow cytometry, the cells were induced to oligodendrocytes using a special differentiation medium. Finally, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), immunocytochemistry, and real-time polymerase chain reaction (RT-PCR) techniques were used for the evaluation of differentiated cells. Results: Flow cytometry indicated that hADSCs were CD105- and CD49-positive, but were negative for CD31 and CD45 markers. In addition, immunocytochemistry analysis revealed that a high percent of differentiated cells expressed oligodendrocyte progenitor cells markers [A2B5 and oligodendrocyte transcription factor (Olig2)] which were significantly higher than myelin basic protein (MBP) which is mature oligodendrocytes marker. Moreover, a very low percentage of differentiated cells expressed glial fibrillary acidic protein (GFAP) marker. Finally, real-time reverse transcription PCR analysis confirmed the results of immunocytochemistry. Conclusion: Since hADSCs have the potential to differentiate into multi-lineage cells and due to their additional characteristics such as immunomodulatory and neuroprotective properties, it seems that these cells may be an ideal cell source for oligodendrocytes differentiation.
Collapse
|
27
|
Sonic hedgehog (SHH) signaling improves the angiogenic potential of Wharton's jelly-derived mesenchymal stem cells (WJ-MSC). Stem Cell Res Ther 2017; 8:203. [PMID: 28962669 PMCID: PMC5622478 DOI: 10.1186/s13287-017-0653-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 08/24/2017] [Accepted: 08/29/2017] [Indexed: 02/07/2023] Open
Abstract
Background Wharton’s jelly-derived mesenchymal stem cells (WJ-MSC) show remarkable therapeutic potential to repair tissue upon injury via paracrine signaling by secreting diverse trophic factors that promote angiogenesis. However, the mechanisms and signaling pathways that regulate the induction of these specific factors are still mostly unknown. Emerging evidence suggests that Sonic hedgehog (SHH) plays a central role in angiogenesis and tissue maintenance. However, its contribution to the angiogenic potential of MSC has not been fully addressed. The aim of this work was to characterize the expression of the SHH pathway components in WJ-MSC primary cultures and to evaluate their angiogenic responsiveness to SHH signaling. Methods Primary cell cultures obtained from human umbilical cords were treated with pharmacological modulators of the SHH pathway. We evaluated the modulation of diverse trophic factors in cell lysates, conditioned medium, and functional in vitro assays. In addition, we determined the angiogenic potential of the SHH pathway in the chicken chorioallantoic membrane, an in vivo model. Results Our results show that WJ-MSC express components of the canonical SHH pathway and are activated by its signaling. In fact, we provide evidence of basal autocrine/paracrine SHH signaling in WJ-MSC. SHH pathway stimulation promotes the secretion of angiogenic factors such as activin A, angiogenin, angiopoietin 1, granulocyte-macrophage colony-stimulating factor, matrix metallometallopeptidase -9, and urokinase-type plasminogen activator, enhancing the pro-angiogenic capabilities of WJ-MSC both in vitro and in vivo. Conclusion WJ-MSC are a cell population responsive to SHH pathway stimulation. Basal SHH signaling is in part responsible for the angiogenic inductive properties of WJ-MSC. Overall, exogenous activation of the SHH pathway enhances the angiogenic properties of WJ-MSC, making this cell population an ideal target for treating tissue injury. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0653-8) contains supplementary material, which is available to authorized users.
Collapse
|
28
|
Adult Stem Cells of Orofacial Origin: Current Knowledge and Limitation and Future Trend in Regenerative Medicine. Tissue Eng Regen Med 2017; 14:719-733. [PMID: 30603522 DOI: 10.1007/s13770-017-0078-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/19/2017] [Accepted: 08/04/2017] [Indexed: 12/21/2022] Open
Abstract
Stem cell research is one of the most rapidly expanding field of medicine which provides significant opportunities for therapeutic and regenerative applications. Different types of stem cells have been isolated investigating their accessibility, control of the differentiation pathway and additional immunomodulatory properties. Bulk of the literature focus has been on the study and potential applications of adult stem cells (ASC) because of their low immunogenicity and reduced ethical considerations. This review paper summarizes the basic available literature on different types of ASC with special focus on stem cells from dental and orofacial origin. ASC have been isolated from different sources, however, isolation of ASC from orofacial tissues has provided a novel promising alternative. These cells offer a great potential in the future of therapeutic and regenerative medicine because of their remarkable availability at low cost while allowing minimally invasive isolation procedures. Furthermore, their immunomodulatory and anti-inflammatory potential is of particular interest. However, there are conflicting reports in the literature regarding their particular biology and full clinical potentials. Sound knowledge and higher control over proliferation and differentiation mechanisms are prerequisites for clinical applications of these cells. Therefore, further standardized basic and translational studies are required to increase the reproducibility and reduce the controversies of studies, which in turn facilitate comparison of related literature and enhance further development in the field.
Collapse
|
29
|
Sepulveda H, Aguilar R, Prieto CP, Bustos F, Aedo S, Lattus J, van Zundert B, Palma V, Montecino M. Epigenetic Signatures at the RUNX2-P1 and Sp7 Gene Promoters Control Osteogenic Lineage Commitment of Umbilical Cord-Derived Mesenchymal Stem Cells. J Cell Physiol 2017; 232:2519-2527. [PMID: 27689934 DOI: 10.1002/jcp.25627] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 09/29/2016] [Indexed: 12/16/2023]
Abstract
Wharton's Jelly mesenchymal stem cells (WJ-MSCs) are an attractive potential source of multipotent stem cells for bone tissue replacement therapies. However, the molecular mechanisms involved in their osteogenic conversion are poorly understood. Particularly, epigenetic control operating at the promoter regions of the two master regulators of the osteogenic program, RUNX2/P57 and SP7 has not yet been described in WJ-MSCs. Via quantitative PCR profiling and chromatin immunoprecipitation (ChIP) studies, here we analyze the ability of WJ-MSCs to engage osteoblast lineage. In undifferentiated WJ-MSCs, RUNX2/P57 P1, and SP7 promoters are found deprived of significant levels of the histone post-translational marks that are normally associated with transcriptionally active genes (H3ac, H3K27ac, and H3K4me3). Moreover, the RUNX2 P1 promoter lacks two relevant histone repressive marks (H3K9me3 and H3K27me3). Importantly, RUNX2 P1 promoter is found highly enriched in the H3K4me1 mark, which has been shown recently to mediate gene repression of key regulatory genes. Upon induction of WJ-MSCs osteogenic differentiation, we found that RUNX2/P57, but not SP7 gene expression is strongly activated, in a process that is accompanied by enrichment of activating histone marks (H3K4me3, H3ac, and H3K27ac) at the P1 promoter region. Histone mark analysis showed that SP7 gene promoter is robustly enriched in epigenetic repressive marks that may explain its poor transcriptional response to osteoblast differentiating media. Together, these results point to critical regulatory steps during epigenetic control of WJ-MSCs osteogenic lineage commitment that are relevant for future applications in regenerative medicine. J. Cell. Physiol. 232: 2519-2527, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hugo Sepulveda
- Center for Biomedical Research, Universidad Andres Bello, Santiago, Chile
- FONDAP Center for Genome Regulation, Santiago, Chile
| | - Rodrigo Aguilar
- Center for Biomedical Research, Universidad Andres Bello, Santiago, Chile
- FONDAP Center for Genome Regulation, Santiago, Chile
| | - Catalina P Prieto
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Santiago, Chile
| | - Francisco Bustos
- FONDAP Center for Genome Regulation, Santiago, Chile
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Santiago, Chile
| | - Sócrates Aedo
- Faculty of Medicine, Department of Obstetrics and Gynecology, Campus Oriente, University of Chile, Santiago, Chile
| | - José Lattus
- Faculty of Medicine, Department of Obstetrics and Gynecology, Campus Oriente, University of Chile, Santiago, Chile
| | | | - Veronica Palma
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Santiago, Chile
| | - Martin Montecino
- Center for Biomedical Research, Universidad Andres Bello, Santiago, Chile
- FONDAP Center for Genome Regulation, Santiago, Chile
| |
Collapse
|
30
|
Dehghani-Soltani S, Shojaee M, Jalalkamali M, Babaee A, Nematollahi-Mahani SN. Effects of light emitting diode irradiation on neural differentiation of human umbilical cord-derived mesenchymal cells. Sci Rep 2017; 7:9976. [PMID: 28855704 PMCID: PMC5577274 DOI: 10.1038/s41598-017-10655-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 08/04/2017] [Indexed: 12/15/2022] Open
Abstract
Recently, light emitting diodes (LEDs) have been introduced as a potential physical factor for proliferation and differentiation of various stem cells. Among the mesenchymal stem cells human umbilical cord matrix-derived mesenchymal (hUCM) cells are easily propagated in the laboratory and their low immunogenicity make them more appropriate for regenerative medicine procedures. We aimed at this study to evaluate the effect of red and green light emitted from LED on the neural lineage differentiation of hUCM cells in the presence or absence of retinoic acid (RA). Harvested hUCM cells exhibited mesenchymal and stemness properties. Irradiation of these cells by green and red LED with or without RA pre-treatment successfully differentiated them into neural lineage when the morphology of the induced cells, gene expression pattern (nestin, β-tubulin III and Olig2) and protein synthesis (anti-nestin, anti-β-tubulin III, anti-GFAP and anti-O4 antibodies) was evaluated. These data point for the first time to the fact that LED irradiation and optogenetic technology may be applied for neural differentiation and neuronal repair in regenerative medicine.
Collapse
Affiliation(s)
- Samereh Dehghani-Soltani
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Mahshid Jalalkamali
- Semiconductors Group, Photonics Research Center, Graduate University of Advanced Technology, Kerman, Iran
| | - Abdolreza Babaee
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | |
Collapse
|
31
|
Deriving vascular smooth muscle cells from mesenchymal stromal cells: Evolving differentiation strategies and current understanding of their mechanisms. Biomaterials 2017; 145:9-22. [PMID: 28843066 DOI: 10.1016/j.biomaterials.2017.08.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/07/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
Vascular smooth muscle cells (VSMCs) play essential roles in regulating blood vessel form and function. Regeneration of functional vascular smooth muscle tissue to repair vascular diseases is an area of intense research in tissue engineering and regenerative medicine. For functional vascular smooth muscle tissue regeneration to become a practical therapy over the next decade, the field will need to have access to VSMC sources that are effective, robust and safe. While pluripotent stem cells hold good future promise to this end, more immediate translation is expected to come from approaches that generate functional VSMCs from adult sources of multipotent adipose-derived and bone marrow-derived mesenchymal stromal cells (ASCs and BMSCs). The research to this end is extensive and is dominated by studies relating to classical biochemical signalling molecules used to induce differentiation of ASCs and BMSCs. However, prolonged use of the biochemical induction factors is costly and can cause potential endotoxin contamination in the culture. Over recent years several non-traditional differentiation approaches have been devised to mimic defined aspects of the native micro-environment in which VSMCs reside to contribute to the differentiation of VSMC-like cells from ASCs and BMSCs. In this review, the promises and limitations of several non-traditional culture approaches (e.g., co-culture, biomechanical, and biomaterial stimuli) targeting VSMC differentiation are discussed. The extensive crosstalk between the underlying signalling cascades are delineated and put into a translational context. It is expected that this review will not only provide significant insight into VSMC differentiation strategies for vascular smooth muscle tissue engineering applications, but will also highlight the fundamental importance of engineering the cellular microenvironment on multiple scales (with consideration of different combinatorial pathways) in order to direct cell differentiation fate and obtain cells of a desired and stable phenotype. These strategies may ultimately be applied to different sources of stem cells in the future for a range of biomaterial and tissue engineering disciplines.
Collapse
|
32
|
Huang M, Qing Y, Shi Q, Cao Y, Song K. miR-342-3p elevates osteogenic differentiation of umbilical cord mesenchymal stem cells via inhibiting Sufu in vitro. Biochem Biophys Res Commun 2017; 491:571-577. [PMID: 28765042 DOI: 10.1016/j.bbrc.2017.07.163] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 07/28/2017] [Indexed: 02/07/2023]
Abstract
Human umbilical mesenchymal stem cells (UCMSCs) have been wildly used in tissue engineering field as a promising source because of their unlimited and noninvasive origin. To selectively induce osteogenic differentiation of UCMSCs, it's imperative to understand the regulatory molecular mechanism underlying the process of how these cells switch into osteogenic lineage path. We previously showed enhanced sonic hedgehog (Shh) signaling pathway upon osteogenic induction in mesenchymal stem cells. In this study, miRNA-seq analysis revealed substantial Shh-dependent expression of distinct miRNAs, including miR-342-3p, during ostogenesis. RT-qPCR confirmed that miR-342-3p was increased at a greater level when Shh signaling pathway was activated by N-terminal of Shh ligand compared with osteogenic induction alone, in contrast to the decreasing of suppressor-of-fused protein (Sufu). Consistently, transient overexpressing miR342-3p in UCMSCs via miR-342-3p mimics dramatically decreased Sufu, a suppressor of Gli, while osteogenic markers, including alkaline phosphate and osteocalcin, were upregulated during osteogenic induction, indicating that miR-342-3p might be involved in osteogenesis through the Shh signaling pathway. In conclusion, this study showed the potential of miR-342-3p as a therapeutic target to promote bone regeneration by modulating expression of Sufu in UCMSCs.
Collapse
Affiliation(s)
- Mengqi Huang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Qing
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Shi
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingguang Cao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Song
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
33
|
Adipose tissue-derived stromal cells (ADSC) express oligodendrocyte and myelin markers, but they do not function as oligodendrocytes. Histochem Cell Biol 2017. [PMID: 28620864 DOI: 10.1007/s00418-017-1588-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Mesenchymal cells cultured from the vasculo-stromal fraction of adipose tissue (ADSC) show adult stem cell characteristics and several groups have claimed generating neural cells from them. However, we have observed that many markers commonly used for the identification of neural cells are spontaneously expressed by ADSC in culture. In the present study, we have examined the expression of characteristic oligodendrocyte molecules in cultured ADSC, aiming to test if myelinating cells could be generated from accessible non-neural adult tissues. In basal growth conditions, rat ADSC spontaneously expressed CNPase, MBP, MOG, protein zero, GAP43, Sox10, and Olig2, as shown by immunocytrochemistry and western blot. A small population of cultured ADSC expressed membrane galactocerebroside (O1 antibody), but no cell stained with O4 antibody. RT-PCR analyses showed the expression of CNPase, MBP, DM20, and low levels of Olig2, Sox10, and Sox2 mRNA by rat ADSC. When rat ADSC were treated with combinations of factors commonly used in neural-inducing media (retinoic acid, dbcAMP, EGF, basic FGF, NT3, and/or PDGF), the number of O1-positive cells changed, but in no case, mRNA expression of Sox10 and Olig2 transcription factors approached CNS oligodendrocyte levels. In co-culture with rat dorsal root ganglion neurons, no sign of axonal myelination by rat ADSC was observed. These studies show that the expression of oligodendrocyte traits by cultured ADSC is not a proof of functional competence as oligodendroglia and suggest that in culture conditions, ADSC acquire intermediate, uncommitted phenotypes.
Collapse
|
34
|
Amelioration of experimental autoimmune encephalomyelitis through transplantation of placental derived mesenchymal stem cells. Sci Rep 2017; 7:41837. [PMID: 28186117 PMCID: PMC5301256 DOI: 10.1038/srep41837] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/29/2016] [Indexed: 12/16/2022] Open
Abstract
Placental derived mesenchymal stem cells (PMSCs) have been suggested as a possible source of cells to treat multiple sclerosis (MS) due to their immunomodulatory functions, lack of ethical concerns, and potential to differentiate into neurons and oligodendrocytes. To investigate whether PMSCs share similar characteristics with embryonic mesenchymal stem cells (EMSCs), and if transplanted PMSCs have the ability to integrate and replace degenerated neural cells, we transplanted rat PMSCs and EMSCs into the central nervous system (CNS) of Lewis rats with experimental autoimmune encephalomyelitis (EAE), an animal model of MS. Our findings demonstrated that transplanted PMSCs, similar to EMSCs, were effective in decreasing infiltrating inflammatory cells, preserving axons, and ameliorating demyelination, thereby improving the neurological functions of animals. Moreover, both PMSCs and EMSCs had the ability to migrate into inflamed tissues and express neural–glial lineage markers. These findings suggest that PMSCs may replace EMSCs as a source of cells in MS stem cell therapy.
Collapse
|
35
|
Luo L, Chen WJ, Yin JQ, Xu RX. EID3 directly associates with DNMT3A during transdifferentiation of human umbilical cord mesenchymal stem cells to NPC-like cells. Sci Rep 2017; 7:40463. [PMID: 28074931 PMCID: PMC5225425 DOI: 10.1038/srep40463] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/06/2016] [Indexed: 12/20/2022] Open
Abstract
There has been recently been increased interest in the plasticity of human umbilical cord mesenchymal stem cells (UMSCs) and their potential in the treatment of neurological disorders. In this study, UMSCs were transdifferentiated into neural stem-like cells (uNSCL), these cells grow in neurosphere-like structures and express high levels of NSCs markers. Epigenetics-related gene screening was here used to assess the relationship between E1A-like inhibitor of differentiation 3 (EID3), a p300 inhibitor, and DNA methyltransferase 3 A (DNMT3A) during the transdifferentiation of UMSCs into uNSCL in vitro. Before transdifferentiation of UMSCs into uNSCLs, high levels of EID3 and low levels of DNMT3A were detected; after transdifferentiation, low levels of EID3 and high levels of DNMT3A were detected. The current work showed that EID3 and DNMT3A co-localized in cell nuclei and EID3 interacted directly with DNMT3A in uNSCL. In summary, these results suggest that DNMT3A is probably directly regulated by EID3 during UMSC transdifferentiation into uNSCLs. These findings indicated a novel mechanism by which EID3, a p300 acetyltransferase inhibitor, could directly affect DNMT3A, this enzyme possesses dual methylation and demethylation abilities. These studies may be helpful for understanding a complex regulation mode of DNMT3A, which is a unique member of the methyltransferase family.
Collapse
Affiliation(s)
- Liang Luo
- Bayi Brain Hospital, General Hospital of PLA Army, Southern Medical University, Beijing 100700, P. R. China.,Stem Cell Research Center, Neurosurgery Institute of Beijing Military Region, Beijing 100700, P. R. China
| | - Wen-Jin Chen
- Bayi Brain Hospital, General Hospital of PLA Army, Southern Medical University, Beijing 100700, P. R. China.,Stem Cell Research Center, Neurosurgery Institute of Beijing Military Region, Beijing 100700, P. R. China
| | - James Q Yin
- Bayi Brain Hospital, General Hospital of PLA Army, Southern Medical University, Beijing 100700, P. R. China.,Stem Cell Research Center, Neurosurgery Institute of Beijing Military Region, Beijing 100700, P. R. China
| | - Ru-Xiang Xu
- Bayi Brain Hospital, General Hospital of PLA Army, Southern Medical University, Beijing 100700, P. R. China.,Stem Cell Research Center, Neurosurgery Institute of Beijing Military Region, Beijing 100700, P. R. China
| |
Collapse
|
36
|
Gao L, Zhao M, Li P, Kong J, Liu Z, Chen Y, Huang R, Chu J, Quan J, Zeng R. Glycogen synthase kinase 3 (GSK3)-inhibitor SB216763 promotes the conversion of human umbilical cord mesenchymal stem cells into neural precursors in adherent culture. Hum Cell 2017; 30:11-22. [PMID: 27604750 DOI: 10.1007/s13577-016-0146-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 08/26/2016] [Indexed: 02/07/2023]
Abstract
The ability to generate neural progenitor cells from human umbilical cord mesenchymal stem cells (hUC-MSCs) has provided an option to treat neurodegenerative diseases. To establish a method for this purpose, we characterized the early neural markers of hUC-MSCs-derived cells under different conditions. We found that neither the elimination of signals for alternative fate nor N2 supplement was sufficient to differentiate hUC-MSCs into neural precursor cells, but the GSK3 inhibitor SB216763 could promote an efficient neural commitment of hUC-MSCs. The results indicated that Wnt/β-catenin might play an important role during the early neural differentiation of hUC-MSCs. Here, we report a method for hUC-MSCs to commit efficiently into a neural fate within a short period of time. This protocol provides an efficient method for hUC-MSCs-based neural regeneration.
Collapse
Affiliation(s)
- Liyang Gao
- Stem Cell Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| | - Mingyan Zhao
- Stem Cell Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Peng Li
- Stem Cell Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Junchao Kong
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhijun Liu
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yonghua Chen
- Department of Pathology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Rui Huang
- Stem Cell Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaqi Chu
- Stem Cell Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Juanhua Quan
- Department of Gastroenterology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Rong Zeng
- Department of Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
37
|
Mueller M, Wolfs TGA, Schoeberlein A, Gavilanes AWD, Surbek D, Kramer BW. Mesenchymal stem/stromal cells-a key mediator for regeneration after perinatal morbidity? Mol Cell Pediatr 2016; 3:6. [PMID: 26869264 PMCID: PMC4751100 DOI: 10.1186/s40348-016-0034-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
Perinatal complications in both term- and preterm-born infants are a leading cause of neonatal morbidities and mortality. Infants face different challenges in the neonatal intensive care unit with long-term morbidities such as perinatal brain injury and bronchopulmonary dysplasia being particularly devastating. While advances in perinatal medicine have improved our understanding of the pathogenesis, effective therapies to prevent and/or reduce the severity of these disorders are still lacking. The potential of mesenchymal stem/stromal cell (MSC) therapy has emerged during the last two decades, and an increasing effort is conducted to address brain- and lung-related morbidities in neonates at risk. Various studies support the notion that MSCs have protective effects. MSCs are an easy source and may be readily available after birth in a clinical setting. MSCs' mechanisms of action are diverse, including migration and homing, release of growth factors and immunomodulation, and the potential to replace injured cells. Here, we review the pathophysiology of perinatally acquired brain and lung injuries and focus on MSCs as potential candidates for therapeutic strategies summarizing preclinical and clinical evidence.
Collapse
Affiliation(s)
- Martin Mueller
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.
- Department of Obstetrics and Gynecology, University Hospital Bern and Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Tim G A Wolfs
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands.
- School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.
| | - Andreina Schoeberlein
- Department of Obstetrics and Gynecology, University Hospital Bern and Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Antonio W D Gavilanes
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands.
- Institute of Biomedicine, Facultad de Ciencias Médicas, Universidad Católica de Santiago de Guayaquil, Guayaquil, Ecuador.
- Department of Neuropsychology, Division Neuroscience, School of Mental Health and neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.
| | - Daniel Surbek
- Department of Obstetrics and Gynecology, University Hospital Bern and Department of Clinical Research, University of Bern, Bern, Switzerland.
| | - Boris W Kramer
- Department of Pediatrics, Maastricht University Medical Center (MUMC), Maastricht, The Netherlands.
- School of Oncology and Developmental Biology (GROW), Maastricht University, Maastricht, The Netherlands.
- Department of Neuropsychology, Division Neuroscience, School of Mental Health and neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
38
|
Takase H, Washida K, Hayakawa K, Arai K, Wang X, Lo EH, Lok J. Oligodendrogenesis after traumatic brain injury. Behav Brain Res 2016; 340:205-211. [PMID: 27829126 DOI: 10.1016/j.bbr.2016.10.042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/20/2016] [Accepted: 10/21/2016] [Indexed: 01/14/2023]
Abstract
White matter injury is an important contributor to long term motor and cognitive dysfunction after traumatic brain injury. During brain trauma, acceleration, deceleration, torsion, and compression forces often cause direct damage to the axon tracts, and pathways that are triggered by the initial injury can trigger molecular events that result in secondary axon degeneration. White matter injury is often associated with altered mental status, memory deficits, motor or autonomic dysfunction, and contribute to the development of chronic neurodegenerative diseases. The presence and proper functioning of oligodendrocyte precursor cells offer the potential for repair and recovery of injured white matter. The process of the proliferation, maturation of oligodendrocyte precursor cells and their migration to the site of injury to replace injured or lost oligodendrocytes is know as oligodendrogenesis. The process of oligodendrogenesis, as well as the interaction of oligodendrocyte precursor cells with other elements of the neurovascular unit, will be discussed in this review.
Collapse
Affiliation(s)
- Hajime Takase
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurosurgery, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kazuo Washida
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States; Division of Neurology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Ken Arai
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Xiaoying Wang
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Eng H Lo
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Radiology, Massachusetts General Hospital, Boston, MA, United States; Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Josephine Lok
- Neuroprotection Research Laboratory, Massachusetts General Hospital, Charlestown, MA, United States; Department of Pediatrics, Massachusetts General Hospital, Boston, MA, United States.
| |
Collapse
|
39
|
Talwadekar M, Fernandes S, Kale V, Limaye L. Valproic acid enhances the neural differentiation of human placenta derived-mesenchymal stem cellsin vitro. J Tissue Eng Regen Med 2016; 11:3111-3123. [DOI: 10.1002/term.2219] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Manasi Talwadekar
- Stem Cell Laboratory, National Centre for Cell Science, NCCS Complex; University of Pune Campus; Ganeshkhind Pune India
| | - Sophia Fernandes
- Stem Cell Laboratory, National Centre for Cell Science, NCCS Complex; University of Pune Campus; Ganeshkhind Pune India
| | - Vaijayanti Kale
- Stem Cell Laboratory, National Centre for Cell Science, NCCS Complex; University of Pune Campus; Ganeshkhind Pune India
| | - Lalita Limaye
- Stem Cell Laboratory, National Centre for Cell Science, NCCS Complex; University of Pune Campus; Ganeshkhind Pune India
| |
Collapse
|
40
|
Mukai T, Nagamura-Inoue T, Shimazu T, Mori Y, Takahashi A, Tsunoda H, Yamaguchi S, Tojo A. Neurosphere formation enhances the neurogenic differentiation potential and migratory ability of umbilical cord-mesenchymal stromal cells. Cytotherapy 2016; 18:229-41. [PMID: 26794714 DOI: 10.1016/j.jcyt.2015.10.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 10/02/2015] [Accepted: 10/24/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND AIMS The human umbilical cord (UC) is a rich source of mesenchymal stromal cells (MSCs), which have been reported to have multi-lineage potential. The objectives of this study were to investigate the characteristics and capacity of UC-MSC neurosphere formation and whether this event enhances the propensity of UC-MSCs to undergo neural differentiation. METHODS UC-MSCs were collected by the improved explant method. UC-MSCs and neurosphere-forming UC-MSCs (UC-MSC-neurospheres) were induced to undergo neurogenic differentiation, the latter of which were induced by suspension culturing in the presence of epidermal growth factor and basic fibroblast growth factor. The differentiation and migratory capacities of the individual cultures were then compared on the basis of the expression of neural markers, as measured by immunocytochemistry, immunoblotting and quantitative real-time polymerase chain reaction and transwell assays, respectively. RESULTS Both UC-MSCs and UC-MSC-neurospheres were capable of differentiating into neurogenic cells when cultured in neurogenic differentiation medium. However, pre-conditioned UC-MSC-neurospheres exhibited significantly higher expression of neural markers--including microtubule-associated protein (MAP2), MUSASHI1, glial fibrillary acidic protein (GFAP), and NESTIN--compared with those derived from UC-MSCs directly. Moreover, UC-MSC-neurospheres expressed significantly higher levels of the stemness markers NANOG, KLF4 and OCT4 than did UC-MSCs. Migration assays also revealed that both UC-MSCs and UC-MSC-neurospheres actively migrate toward glucose-depleted cells. CONCLUSIONS Neurogenic differentiation potential probably is greater in UC-MSC-neurospheres than in UC-MSCs. Thus, UC-MSC-neurospheres may serve as a better source of cells for neurogenic regenerative medicine.
Collapse
Affiliation(s)
- Takeo Mukai
- Division of Molecular of Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Tokiko Nagamura-Inoue
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| | - Takahisa Shimazu
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuka Mori
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Atsuko Takahashi
- Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hajime Tsunoda
- Department of Obstetrics, NTT Medical Center Tokyo Hospital, Tokyo, Japan
| | | | - Arinobu Tojo
- Division of Molecular of Therapy, Center for Advanced Medical Research, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Cell Processing and Transfusion, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
41
|
Zhang Q, Nguyen P, Xu Q, Park W, Lee S, Furuhashi A, Le AD. Neural Progenitor-Like Cells Induced from Human Gingiva-Derived Mesenchymal Stem Cells Regulate Myelination of Schwann Cells in Rat Sciatic Nerve Regeneration. Stem Cells Transl Med 2016; 6:458-470. [PMID: 28191764 PMCID: PMC5442816 DOI: 10.5966/sctm.2016-0177] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/28/2016] [Indexed: 12/26/2022] Open
Abstract
Regeneration of peripheral nerve injury remains a major clinical challenge. Recently, mesenchymal stem cells (MSCs) have been considered as potential candidates for peripheral nerve regeneration; however, the underlying mechanisms remain elusive. Here, we show that human gingiva‐derived MSCs (GMSCs) could be directly induced into multipotent NPCs (iNPCs) under minimally manipulated conditions without the introduction of exogenous genes. Using a crush‐injury model of rat sciatic nerve, we demonstrate that GMSCs transplanted to the injury site could differentiate into neuronal cells, whereas iNPCs could differentiate into both neuronal and Schwann cells. After crush injury, iNPCs, compared with GMSCs, displayed superior therapeutic effects on axonal regeneration at both the injury site and the distal segment of the injured sciatic nerve. Mechanistically, transplantation of GMSCs, especially iNPCs, significantly attenuated injury‐triggered increase in the expression of c‐Jun, a transcription factor that functions as a major negative regulator of myelination and plays a central role in dedifferentiation/reprogramming of Schwann cells into a progenitor‐like state. Meanwhile, our results also demonstrate that transplantation of GMSCs and iNPCs consistently increased the expression of Krox‐20/EGR2, a transcription factor that governs the expression of myelin proteins and facilitates myelination. Altogether, our findings suggest that transplantation of GMSCs and iNPCs promotes peripheral nerve repair/regeneration, possibly by promoting remyelination of Schwann cells mediated via the regulation of the antagonistic myelination regulators, c‐Jun and Krox‐20/EGR2. Stem Cells Translational Medicine2017;6:458–470
Collapse
Affiliation(s)
- Qunzhou Zhang
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Phuong Nguyen
- Division of Plastic and Reconstructive Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Qilin Xu
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Wonse Park
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Sumin Lee
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Akihiro Furuhashi
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Anh D. Le
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
- Department of Oral and Maxillofacial Surgery, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
42
|
Villalón H, Peñaloza G, Tuma D. TERAPIA REGENERATIVA EN NEONATOLOGÍA. REVISTA MÉDICA CLÍNICA LAS CONDES 2016. [DOI: 10.1016/j.rmclc.2016.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
43
|
Madill M, Fitzgerald D, O'Connell KE, Dev KK, Shen S, FitzGerald U. In vitro and ex vivo models of multiple sclerosis. Drug Discov Today 2016; 21:1504-1511. [PMID: 27265771 DOI: 10.1016/j.drudis.2016.05.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/20/2016] [Accepted: 05/27/2016] [Indexed: 01/25/2023]
Abstract
Multiple sclerosis (MS) is an autoimmune disorder of the central nervous system (CNS). Current therapies suppress a misdirected myelin-destructive immune response. To combat the progressive, neurodestructive phase of MS, the therapeutic research focus is currently on compounds that might boost the endogenous potential of the brain to remyelinate axons, thereby achieving lesion repair. Here, we describe the testing of fingolimod on cultures of oligodendrocytes (OLs) and organotypic brain slices. We detail the protocols, pros, and cons of these in vitro and ex vivo approaches, along with the potential benefit of exploiting skin-punch biopsies from patients with MS, before concluding with a summary of future developments.
Collapse
Affiliation(s)
- Martin Madill
- Regenerative Medicine Institute (REMEDI), School of Medicine and School of Natural Sciences, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Denise Fitzgerald
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast, UK
| | - Kara E O'Connell
- Drug Development, School of Medicine, Trinity College Dublin, Ireland
| | - Kumlesh K Dev
- Drug Development, School of Medicine, Trinity College Dublin, Ireland
| | - Sanbing Shen
- Regenerative Medicine Institute (REMEDI), School of Medicine and School of Natural Sciences, National University of Ireland (NUI) Galway, Galway, Ireland
| | - Una FitzGerald
- Galway Neuroscience Centre, School of Natural Sciences, National University of Ireland, Galway, Ireland.
| |
Collapse
|
44
|
Zhao L, Feng Y, Chen X, Yuan J, Liu X, Chen Y, Zhao Y, Liu P, Li Y. Effects of IGF-1 on neural differentiation of human umbilical cord derived mesenchymal stem cells. Life Sci 2016; 151:93-101. [DOI: 10.1016/j.lfs.2016.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 02/28/2016] [Accepted: 03/01/2016] [Indexed: 12/12/2022]
|
45
|
Magnetic induction heating of superparamagnetic nanoparticles during rewarming augments the recovery of hUCM-MSCs cryopreserved by vitrification. Acta Biomater 2016; 33:264-74. [PMID: 26802443 DOI: 10.1016/j.actbio.2016.01.026] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 12/24/2015] [Accepted: 01/19/2016] [Indexed: 01/03/2023]
Abstract
Cryopreservation by vitrification has been recognized as a promising strategy for long-term banking of living cells. However, the difficulty to generate a fast enough heating rate to minimize devitrification and recrystallization-induced intracellular ice formation during rewarming is one of the major obstacles to successful vitrification. We propose to overcome this hurdle by utilizing magnetic induction heating (MIH) of magnetic nanoparticles to enhance rewarming. In this study, superparamagnetic (SPM) Fe3O4 nanoparticles were synthesized by a chemical coprecipitation method. We successfully applied the MIH of Fe3O4 nanoparticles for rewarming human umbilical cord matrix mesenchymal stem cells (hUCM-MSCs) cryopreserved by vitrification. Our results show that extracellular Fe3O4 nanoparticles with MIH may efficiently suppress devitrification and/or recrystallization during rewarming and significantly improve the survival of vitrified cells. We further optimized the concentration of Fe3O4 nanoparticles and the current of an alternating current (AC) magnetic field for generating the MIH to maximize cell viability. Our results indicate that MIH in an AC magnetic field with 0.05% (w/v) Fe3O4 nanoparticles significantly facilitates rewarming and improves the cryopreservation outcome of hUCM-MSCs by vitrification. The application of MIH of SPM nanoparticles to achieve rapid and spatially homogeneous heating is a promising strategy for enhanced cryopreservation of stem cells by vitrification. STATEMENT OF SIGNIFICANCE Here we report the successful synthesis and application of Fe3O4 nanoparticles for magnetic induction heating (MIH) to enhance rewarming of vitrification-cryopreserved human umbilical cord matrix mesenchymal stem cells (hUCM-MSCs). We found that MIH-enhanced rewarming greatly improves the survival of vitrification-cryopreserved hUCM-MSCs. Moreover, the hUCM-MSCs retain their intact stemness and multilineage potential of differentiation post cryopreservation by vitrification with the MIH-enhanced rewarming. Therefore, the novel MIH-enhanced cell vitrification is valuable to facilitate the long-term storage of hUCM-MSCs and possibly many other important cells to meet their ever-increasing demand by the burgeoning cell-based medicine.
Collapse
|
46
|
Zou Q, Wu M, Zhong L, Fan Z, Zhang B, Chen Q, Ma F. Development of a Xeno-Free Feeder-Layer System from Human Umbilical Cord Mesenchymal Stem Cells for Prolonged Expansion of Human Induced Pluripotent Stem Cells in Culture. PLoS One 2016; 11:e0149023. [PMID: 26882313 PMCID: PMC4755601 DOI: 10.1371/journal.pone.0149023] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/26/2016] [Indexed: 02/06/2023] Open
Abstract
Various feeder layers have been extensively applied to support the prolonged growth of human pluripotent stem cells (hPSCs) for in vitro cultures. Among them, mouse embryonic fibroblast (MEF) and mouse fibroblast cell line (SNL) are most commonly used feeder cells for hPSCs culture. However, these feeder layers from animal usually cause immunogenic contaminations, which compromises the potential of hPSCs in clinical applications. In the present study, we tested human umbilical cord mesenchymal stem cells (hUC-MSCs) as a potent xeno-free feeder system for maintaining human induced pluripotent stem cells (hiPSCs). The hUC-MSCs showed characteristics of MSCs in xeno-free culture condition. On the mitomycin-treated hUC-MSCs feeder, hiPSCs maintained the features of undifferentiated human embryonic stem cells (hESCs), such as low efficiency of spontaneous differentiation, stable expression of stemness markers, maintenance of normal karyotypes, in vitro pluripotency and in vivo ability to form teratomas, even after a prolonged culture of more than 30 passages. Our study indicates that the xeno-free culture system may be a good candidate for growth and expansion of hiPSCs as the stepping stone for stem cell research to further develop better and safer stem cells.
Collapse
Affiliation(s)
- Qing Zou
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
- Center for Stem Cell Research & Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
| | - Mingjun Wu
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
| | - Liwu Zhong
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
| | - Zhaoxin Fan
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
| | - Bo Zhang
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
| | - Qiang Chen
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
- Center for Stem Cell Research & Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
- * E-mail: (FM); (QC)
| | - Feng Ma
- Research Center for Stem Cell and Regenerative Medicine, Sichuan Neo-life Stem Cell Biotech INC., Chengdu, Sichuan, China
- Center for Stem Cell Research & Application, Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu, Sichuan, China
- State Key Laboratory of Experimental Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- * E-mail: (FM); (QC)
| |
Collapse
|
47
|
Lourenço T, Paes de Faria J, Bippes CA, Maia J, Lopes-da-Silva JA, Relvas JB, Grãos M. Modulation of oligodendrocyte differentiation and maturation by combined biochemical and mechanical cues. Sci Rep 2016; 6:21563. [PMID: 26879561 PMCID: PMC4754901 DOI: 10.1038/srep21563] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 01/25/2016] [Indexed: 01/17/2023] Open
Abstract
Extracellular matrix (ECM) proteins play a key role during oligodendrogenesis. While fibronectin (FN) is involved in the maintenance and proliferation of oligodendrocyte progenitor cells (OPCs), merosin (MN) promotes differentiation into oligodendrocytes (OLs). Mechanical properties of the ECM also seem to affect OL differentiation, hence this study aimed to clarify the impact of combined biophysical and biochemical elements during oligodendrocyte differentiation and maturation using synthetic elastic polymeric ECM-like substrates. CG-4 cells presented OPC- or OL-like morphology in response to brain-compliant substrates functionalised with FN or MN, respectively. The expression of the differentiation and maturation markers myelin basic protein — MBP — and proteolipid protein — PLP — (respectively) by primary rat oligodendrocytes was enhanced in presence of MN, but only on brain-compliant conditions, considering the distribution (MBP) or amount (PLP) of the protein. It was also observed that maturation of OLs was attained earlier (by assessing PLP expression) by cells differentiated on MN-functionalised brain-compliant substrates than on standard culture conditions. Moreover, the combination of MN and substrate compliance enhanced the maturation and morphological complexity of OLs. Considering the distinct degrees of stiffness tested ranging within those of the central nervous system, our results indicate that 6.5 kPa is the most suitable rigidity for oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Tânia Lourenço
- Biocant, Technology Transfer Association, Cantanhede, Portugal.,Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Joana Paes de Faria
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | | | - João Maia
- Chemical Engineering Department, Faculty of Science and Technology, University of Coimbra, Coimbra, Portugal
| | | | - João B Relvas
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
| | - Mário Grãos
- Biocant, Technology Transfer Association, Cantanhede, Portugal.,Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
48
|
Shahbazi A, Safa M, Alikarami F, Kargozar S, Asadi MH, Joghataei MT, Soleimani M. Rapid Induction of Neural Differentiation in Human Umbilical Cord Matrix Mesenchymal Stem Cells by cAMP-elevating Agents. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2016; 5:167-177. [PMID: 27942503 PMCID: PMC5125369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/22/2016] [Indexed: 11/15/2022]
Abstract
Human umbilical cord matrix (hUCM) is considered as a promising source of mesenchymal stem cells (MSCs) due to several advantages over other tissues. The potential of neural differentiation of hUCM-MSCs is of great interest in the context of treating neurodegenerative diseases. In recent years, considerable efforts have been made to establish in vitro conditions for improving the differentiation of hUCM-MSCs toward neuronal cells. In the present study, we evaluated the neural differentiation potential of hUCM-MSCs in the presence of cAMP-elevating agents forskolin and 3-isobutyl-1-methylxanthine (IBMX). hUCM-MSCs were isolated from fetal umbilical cord and characterized by flow cytometry analysis for mesenchymal specific markers. Mesodermal differentiation potential was assessed through selective media with lineage-specific induction factors. For assessment of neural differentiation, cells were cultured in the presence of cAMP-elevating agents for 8 and 24 h. The neuronal differentiated MSCs were characterized for neuronal specific markers by immunocytochemistry and western blotting. Isolated hUCM-MSCs were found positive for mesenchymal markers (CD73, CD90, and CD105) while negative for hematopoietic markers (CD34 and CD45) .Following neural induction, most cells represented neural-like cells morphology. Neural markers including β-tubulin III (Tuj-1), neuron-specific enolase (NSE), microtubule-associated protein-2 (MAP-2) and nestin were expressed in treated cells with respect to control group. The astrocyte specific marker, glial fibrillary acidic protein (GFAP) was also shown by immunofluorescence in treated cells. (These findings demonstrate that hUCM-MSCs have the ability to rapidly differentiate into neural cell types of neuron-like cells and astrocytes by cAMP-elevating agents without the presence of growth factors.
Collapse
Affiliation(s)
- Atefeh Shahbazi
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Majid Safa
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.,Department of Hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.,Corresponding author: Cellular and Molecular Research Center and Department of Hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran. E-mail:
| | - Fatemeh Alikarami
- Department of Hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Saeid Kargozar
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Hossein Asadi
- Department of Anatomy, School of Medicine, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | | | - Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
49
|
Karaöz E, İnci Ç. Umbilical Cord Tissue and Wharton’s Jelly Mesenchymal Stem Cells Properties and Therapeutic Potentials. PERINATAL TISSUE-DERIVED STEM CELLS 2016. [DOI: 10.1007/978-3-319-46410-7_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
50
|
Optimized Protocol for Isolation of Multipotent Mesenchymal Stromal Cells from Human Umbilical Cord. Bull Exp Biol Med 2015; 160:148-54. [DOI: 10.1007/s10517-015-3116-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Indexed: 12/12/2022]
|