1
|
Liu S, Chu X, Reiter JL, Yu X, Fang F, McGuire P, Gao H, Liu Y, Wan J, Wang Y. Dynamic chromatin accessibility and transcriptome changes following PDGF-BB treatment of bone-marrow derived mesenchymal stem cells. BMC Genomics 2024; 25:962. [PMID: 39407135 PMCID: PMC11476831 DOI: 10.1186/s12864-024-10861-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/03/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are multipotent stem cells that are under investigation for use in clinical trials because they are capable of self-renewal and differentiating into different cell types under defined conditions. Nonetheless, the therapeutic effects of MSCs have been constrained by low engraftment rates, cell fusion, and cell survival. Various strategies have been explored to improve the therapeutic efficacy of MSCs, with platelet-derived growth factor (PDGF)-BB emerging as a promising candidate. To enhance our comprehension of the impact of PDGF-BB on the gene expression profile and chromosomal accessibility of MSCs, RNA-sequencing and analysis of chromatin accessibility profiles were conducted on three human primary MSCs in culture, both with and without stimulation by PDGF-BB. RESULTS Integrative analysis of gene expression and chromatin accessibility demonstrated that PDGF-BB treatment modified the chromatin accessibility landscape, marking regions for activation or repression through the AP-1 family transcription factors TEAD, CEBP, and RUNX2. These changes in AP-1 transcription factor expression, in turn, led to cell proliferation and differentiation potential towards osteoblasts, adipocytes, or chondrocytes. The degree of MSC differentiation varies among cells isolated from different donors. The presence of an enrichment of exosome-related genes is also noted among all the differentially expressed genes. CONCLUSIONS In conclusion, the observed changes in AP-1 transcription factor expression not only induced cellular proliferation and differentiation, but also revealed variations in the degree of MSC differentiation based on donor-specific differences. Moreover, the enrichment of exosome-related genes among differentially expressed genes suggests a potential significant role for PDGF-BB in facilitating intercellular communication.
Collapse
Affiliation(s)
- Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiaona Chu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jill L Reiter
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xuhong Yu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fang Fang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Patrick McGuire
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Yue Wang
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
2
|
Gornostaeva AN, Buravkova LB. Changes Induced by Inflammatory-Activated Immune Cell Microenvironment in the Paracrine Profile of MSC. Bull Exp Biol Med 2023; 174:544-548. [PMID: 36894814 DOI: 10.1007/s10517-023-05745-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Indexed: 03/11/2023]
Abstract
We studied the influence of activated innate and adaptive immune cells on the production of growth factors by human adipose tissue multipotent mesenchymal stromal cells (MSC). MSC showed immunosuppressive properties in vitro: decreased activation and proliferation of stimulated immune cells. T-cell interaction with MSC resulted with increased secretion of EGF, PDGF-AB/BB, FGF-2, and VEGF growth factors. Co-culturing with natural killer cells also stimulated TGFα production. The intensity of the effect varied depending on the type of immune cells. Natural killer caused a more significant increase in PDGF-AB/BB and FGF-2 secretion, while VEGF secretion increased stronger after co-culturing with T cells. The obtained data indicate the possibility of increasing reparative potential of MSC under the influence of inflammatory microenvironment.
Collapse
Affiliation(s)
- A N Gornostaeva
- Institute of Biomedical Problems, State Research Center, Russian Academy of Sciences, Moscow, Russia.
| | - L B Buravkova
- Institute of Biomedical Problems, State Research Center, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
3
|
Mabotuwana NS, Rech L, Lim J, Hardy SA, Murtha LA, Rainer PP, Boyle AJ. Paracrine Factors Released by Stem Cells of Mesenchymal Origin and their Effects in Cardiovascular Disease: A Systematic Review of Pre-clinical Studies. Stem Cell Rev Rep 2022; 18:2606-2628. [PMID: 35896860 PMCID: PMC9622561 DOI: 10.1007/s12015-022-10429-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2022] [Indexed: 11/30/2022]
Abstract
Mesenchymal stem cell (MSC) therapy has gained significant traction in the context of cardiovascular repair, and have been proposed to exert their regenerative effects via the secretion of paracrine factors. In this systematic review, we examined the literature and consolidated available evidence for the "paracrine hypothesis". Two Ovid SP databases were searched using a strategy encompassing paracrine mediated MSC therapy in the context of ischemic heart disease. This yielded 86 articles which met the selection criteria for inclusion in this study. We found that the MSCs utilized in these articles were primarily derived from bone marrow, cardiac tissue, and adipose tissue. We identified 234 individual protective factors across these studies, including VEGF, HGF, and FGF2; which are proposed to exert their effects in a paracrine manner. The data collated in this systematic review identifies secreted paracrine factors that could decrease apoptosis, and increase angiogenesis, cell proliferation, and cell viability. These included studies have also demonstrated that the administration of MSCs and indirectly, their secreted factors can reduce infarct size, and improve left ventricular ejection fraction, contractility, compliance, and vessel density. Furthering our understanding of the way these factors mediate repair could lead to the identification of therapeutic targets for cardiac regeneration.
Collapse
Affiliation(s)
- Nishani S Mabotuwana
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Lavinia Rech
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
- Department of Cardiac Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Joyce Lim
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, NSW, Australia
| | - Sean A Hardy
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Lucy A Murtha
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia
| | - Peter P Rainer
- Department of Internal Medicine, Division of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Andrew J Boyle
- College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia.
- Hunter Medical Research Institute, Lot 1, Kookaburra Circuit, Newcastle, NSW, 2305, Australia.
- Department of Cardiovascular Medicine, John Hunter Hospital, Newcastle, NSW, Australia.
| |
Collapse
|
4
|
Zhang H, Liu G, Mao X, Yang L, Wang B, Yuan X. LncRNA MEG3 induces endothelial differentiation of mouse derived adipose-derived stem cells by targeting MiR-145-5p/KLF4. Mol Biol Rep 2022; 49:8495-8505. [PMID: 35802277 DOI: 10.1007/s11033-022-07671-z] [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: 01/19/2022] [Accepted: 06/01/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND The present study aimed to investigate the mechanisms through which long non-coding RNA (lncRNA) maternally expressed 3 (MEG3) affected the endothelial differentiation of mouse derived adipose-derived stem cells (ADSCs). MATERIALS AND METHODS ADSCs were isolated and identified by specific surface marker detection. The effects of lncRNA MEG3 on endothelial differentiation of ADSCs were also detected via quantitative PCR, western blotting, immunofluorescence and Matrigel angiogenesis assays. In addition, using target gene prediction tools and luciferase reporter assays, the downstream target gene was demonstrated. RESULTS LncRNA MEG3 targeted and reduced the expression levels of microRNA-145-5p (miR-145-5p), which upregulated the expression levels of Krüppel like factor 4 (KLF4), promoting endothelial differentiation of ADSCs. CONCLUSION LncRNA MEG3 induced endothelial differentiation of ADSCs by targeting miR-145-5p/KLF4, which may provide novel insights to illustrate the mechanism of endothelial differentiation of ADSCs.
Collapse
Affiliation(s)
- Hailong Zhang
- Department of Dermatology, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150006, Heilongjiang, People's Republic of China
| | - Gang Liu
- Department of Medicine, Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150006, Heilongjiang, People's Republic of China
| | - Xu Mao
- Department of Health Center, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, Heilongjiang, People's Republic of China
| | - Lei Yang
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, No. 33 of West Dazhi Street, Harbin, 150001, Heilongjiang, People's Republic of China
| | - Bingyu Wang
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, No. 33 of West Dazhi Street, Harbin, 150001, Heilongjiang, People's Republic of China.
| | - Xingxing Yuan
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, No. 33 of West Dazhi Street, Harbin, 150001, Heilongjiang, People's Republic of China.
| |
Collapse
|
5
|
Hu W, Yang C, Guo X, Wu Y, Loh XJ, Li Z, Wu YL, Wu C. Research Advances of Injectable Functional Hydrogel Materials in the Treatment of Myocardial Infarction. Gels 2022; 8:423. [PMID: 35877508 PMCID: PMC9316750 DOI: 10.3390/gels8070423] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 12/10/2022] Open
Abstract
Myocardial infarction (MI) has become one of the serious diseases threatening human life and health. However, traditional treatment methods for MI have some limitations, such as irreversible myocardial necrosis and cardiac dysfunction. Fortunately, recent endeavors have shown that hydrogel materials can effectively prevent negative remodeling of the heart and improve the heart function and long-term prognosis of patients with MI due to their good biocompatibility, mechanical properties, and electrical conductivity. Therefore, this review aims to summarize the research progress of injectable hydrogel in the treatment of MI in recent years and to introduce the rational design of injectable hydrogels in myocardial repair. Finally, the potential challenges and perspectives of injectable hydrogel in this field will be discussed, in order to provide theoretical guidance for the development of new and effective treatment strategies for MI.
Collapse
Affiliation(s)
- Wei Hu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Cui Yang
- School of Medicine, Xiamen University, Xiamen 361003, China;
| | - Xiaodan Guo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Yihong Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore;
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore;
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE) Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| |
Collapse
|
6
|
Receptor tyrosine kinase inhibitors negatively impact on pro-reparative characteristics of human cardiac progenitor cells. Sci Rep 2022; 12:10132. [PMID: 35710779 PMCID: PMC9203790 DOI: 10.1038/s41598-022-13203-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 05/23/2022] [Indexed: 12/21/2022] Open
Abstract
Receptor tyrosine kinase inhibitors improve cancer survival but their cardiotoxicity requires investigation. We investigated these inhibitors’ effects on human cardiac progenitor cells in vitro and rat heart in vivo. We applied imatinib, sunitinib or sorafenib to human cardiac progenitor cells, assessing cell viability, proliferation, stemness, differentiation, growth factor production and second messengers. Alongside, sunitinib effects were assessed in vivo. Inhibitors decreased (p < 0.05) cell viability, at levels equivalent to ‘peak’ (24 h; imatinib: 91.5 ± 0.9%; sunitinib: 83.9 ± 1.8%; sorafenib: 75.0 ± 1.6%) and ‘trough’ (7 days; imatinib: 62.3 ± 6.2%; sunitinib: 86.2 ± 3.5%) clinical plasma levels, compared to control (100% viability). Reduced (p < 0.05) cell cycle activity was seen with imatinib (29.3 ± 4.3% cells in S/G2/M-phases; 50.3 ± 5.1% in control). Expression of PECAM-1, Nkx2.5, Wnt2, linked with cell differentiation, were decreased (p < 0.05) 2, 2 and 6-fold, respectively. Expression of HGF, p38 and Akt1 in cells was reduced (p < 0.05) by sunitinib. Second messenger (p38 and Akt1) blockade affected progenitor cell phenotype, reducing c-kit and growth factor (HGF, EGF) expression. Sunitinib for 9 days (40 mg/kg, i.p.) in adult rats reduced (p < 0.05) cardiac ejection fraction (68 ± 2% vs. baseline (83 ± 1%) and control (84 ± 4%)) and reduced progenitor cell numbers. Receptor tyrosine kinase inhibitors reduce cardiac progenitor cell survival, proliferation, differentiation and reparative growth factor expression.
Collapse
|
7
|
Sanches BDA, Maldarine JS, Vilamaior PSL, Felisbino SL, Carvalho HF, Taboga SR. Stromal cell interplay in prostate development, physiology, and pathological conditions. Prostate 2021; 81:926-937. [PMID: 34254335 DOI: 10.1002/pros.24196] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/18/2021] [Accepted: 06/29/2021] [Indexed: 12/18/2022]
Abstract
Advances in prostatic stroma studies over the past few decades have demonstrated that the stroma not only supports and nourishes the gland's secretory epithelium but also participates in key aspects of morphogenesis, in the prostate's hormonal metabolism, and in the functionality of the secretory epithelium. Furthermore, the stroma is implicated in the onset and progression of prostate cancer through the formation of the so-called reactive stroma, which corresponds to a tumorigenesis-permissive microenvironment. Prostatic stromal cells are interconnected and exchange paracrine signals among themselves in a gland that is highly sensitive to endocrine hormones. There is a growing body of evidence that telocytes, recently detected interstitial cells that are also present in the prostate, are involved in stromal organization, so that their processes form a network of interconnections with both the epithelium and the other stromal cells. The present review provides an update on the different types of prostate stromal cells, their interrelationships and implications for prostate development, physiology and pathological conditions.
Collapse
Affiliation(s)
- Bruno D A Sanches
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Juliana S Maldarine
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Patricia S L Vilamaior
- Department of Biological Sciences, Laboratory of Microscopy and Microanalysis, São Paulo State University-UNESP, São José do Rio Preto, Brazil
| | - Sergio L Felisbino
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
- Institute of Biosciences, São Paulo State University-UNESP, Botucatu, Brazil
| | - Hernandes F Carvalho
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
| | - Sebastião R Taboga
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, Brazil
- Department of Biological Sciences, Laboratory of Microscopy and Microanalysis, São Paulo State University-UNESP, São José do Rio Preto, Brazil
| |
Collapse
|
8
|
Beliën H, Evens L, Hendrikx M, Bito V, Bronckaers A. Combining stem cells in myocardial infarction: The road to superior repair? Med Res Rev 2021; 42:343-373. [PMID: 34114238 DOI: 10.1002/med.21839] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/04/2021] [Accepted: 05/29/2021] [Indexed: 12/25/2022]
Abstract
Myocardial infarction irreversibly destroys millions of cardiomyocytes in the ventricle, making it the leading cause of heart failure worldwide. Over the past two decades, many progenitor and stem cell types were proposed as the ideal candidate to regenerate the heart after injury. The potential of stem cell therapy has been investigated thoroughly in animal and human studies, aiming at cardiac repair by true tissue replacement, by immune modulation, or by the secretion of paracrine factors that stimulate endogenous repair processes. Despite some successful results in animal models, the outcome from clinical trials remains overall disappointing, largely due to the limited stem cell survival and retention after transplantation. Extensive interest was developed regarding the combinational use of stem cells and various priming strategies to improve the efficacy of regenerative cell therapy. In this review, we provide a critical discussion of the different stem cell types investigated in preclinical and clinical studies in the field of cardiac repair. Moreover, we give an update on the potential of stem cell combinations as well as preconditioning and explore the future promises of these novel regenerative strategies.
Collapse
Affiliation(s)
- Hanne Beliën
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Lize Evens
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Marc Hendrikx
- Faculty of Medicine and Life Sciences, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Virginie Bito
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| | - Annelies Bronckaers
- Biomedical Research Institute (BIOMED), Department of Cardio and Organ Systems, UHasselt-Hasselt University, Agoralaan, Diepenbeek, Belgium
| |
Collapse
|
9
|
Parmaksiz M, Elçin AE, Elçin YM. Decellularized Cell Culture ECMs Act as Cell Differentiation Inducers. Stem Cell Rev Rep 2021; 16:569-584. [PMID: 32170583 DOI: 10.1007/s12015-020-09963-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Decellularized tissues and organs have aroused considerable interest for developing functional bio-scaffolds as natural templates in tissue engineering applications. More recently, the use of natural extracellular matrix (ECM) extracted from the in vitro cell cultures for cellular applications have come into question. It is well known that the microenvironment largely defines cellular properties. Thus, we have anticipated that the ECMs of the cells with different potency levels should likely possess different effects on cell cultures. To test this, we have comparatively evaluated the differentiative effects of ECMs derived from the cultures of human somatic dermal fibroblasts, human multipotent bone marrow mesenchymal stem cells, and human induced pluripotent stem cells on somatic dermal fibroblasts. Although challenges remain, the data suggest that the use of cell culture-based extracellular matrices perhaps may be considered as an alternative approach for the differentiation of even somatic cells into other cell types.
Collapse
Affiliation(s)
- Mahmut Parmaksiz
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey
| | - Ayşe Eser Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey
| | - Yaşar Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Ankara, Turkey. .,Biovalda Health Technologies, Inc, Ankara, Turkey.
| |
Collapse
|
10
|
Wu Y, Chang T, Chen W, Wang X, Li J, Chen Y, Yu Y, Shen Z, Yu Q, Zhang Y. Release of VEGF and BMP9 from injectable alginate based composite hydrogel for treatment of myocardial infarction. Bioact Mater 2021; 6:520-528. [PMID: 32995677 PMCID: PMC7492819 DOI: 10.1016/j.bioactmat.2020.08.031] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/21/2020] [Accepted: 08/29/2020] [Indexed: 12/28/2022] Open
Abstract
Myocardial infarction (MI) is one of cardiovascular diseases that pose a serious threat to human health. The pathophysiology of MI is complex and contains several sequential phases including blockage of a coronary artery, necrosis of myocardial cells, inflammation, and myocardial fibrosis. Aiming at the treatment of different stages of MI, in this work, an injectable alginate based composite hydrogel is developed to load vascular endothelial active factor (VEGF) and silk fibroin (SF) microspheres containing bone morphogenetic protein 9 (BMP9) for releasing VEGF and BMP9 to realize their respective functions. The results of in vitro experiments indicate a rapid initial release of VEGF during the first few days and a relatively slow and sustained release of BMP9 for days, facilitating the formation of blood vessels in the early stage and inhibiting myocardial fibrosis in the long-term stage, respectively. Intramyocardial injection of such composite hydrogel into the infarct border zone of mice MI model via multiple points promotes angiogenesis and reduces the infarction size. Taken together, these results indicate that the dual-release of VEGF and BMP9 from the composite hydrogel results in a collaborative effect on the treatment of MI and improvement of heart function, showing a promising potential for cardiac clinical application.
Collapse
Affiliation(s)
- Yong Wu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, PR China
| | - Tianqi Chang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, PR China
| | - Weiqian Chen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, PR China
| | - Xiaoyu Wang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, PR China
| | - Jingjing Li
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, PR China
| | - Yueqiu Chen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, PR China
| | - You Yu
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, PR China
| | - Zhenya Shen
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, PR China
| | - Qian Yu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, PR China
| | - Yanxia Zhang
- Institute for Cardiovascular Science & Department of Cardiovascular Surgery of the First Affiliated Hospital, Medical College, Soochow University, Suzhou, 215000, PR China
| |
Collapse
|
11
|
Nwabo Kamdje AH, Seke Etet PF, Simo Tagne R, Vecchio L, Lukong KE, Krampera M. Tumor Microenvironment Uses a Reversible Reprogramming of Mesenchymal Stromal Cells to Mediate Pro-tumorigenic Effects. Front Cell Dev Biol 2020; 8:545126. [PMID: 33330442 PMCID: PMC7710932 DOI: 10.3389/fcell.2020.545126] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 10/28/2020] [Indexed: 12/13/2022] Open
Abstract
The role of mesenchymal stromal cells (MSCs) in the tumor microenvironment is well described. Available data support that MSCs display anticancer activities, and that their reprogramming by cancer cells in the tumor microenvironment induces their switch toward pro-tumorigenic activities. Here we discuss the recent evidence of pro-tumorigenic effects of stromal cells, in particular (i) MSC support to cancer cells through the metabolic reprogramming necessary to maintain their malignant behavior and stemness, and (ii) MSC role in cancer cell immunosenescence and in the establishment and maintenance of immunosuppression in the tumor microenvironment. We also discuss the mechanisms of tumor microenvironment mediated reprogramming of MSCs, including the effects of hypoxia, tumor stiffness, cancer-promoting cells, and tumor extracellular matrix. Finally, we summarize the emerging strategies for reprogramming tumor MSCs to reactivate anticancer functions of these stromal cells.
Collapse
Affiliation(s)
- Armel H. Nwabo Kamdje
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences (FMBS), University of Ngaoundéré, Ngaoundéré, Cameroon
| | - Paul F. Seke Etet
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences (FMBS), University of Ngaoundéré, Ngaoundéré, Cameroon
- Center for Sustainable Health and Development, Garoua, Cameroon
| | - Richard Simo Tagne
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences (FMBS), University of Ngaoundéré, Ngaoundéré, Cameroon
| | - Lorella Vecchio
- Center for Sustainable Health and Development, Garoua, Cameroon
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Mauro Krampera
- Section of Hematology, Stem Cell Research Laboratory, Department of Medicine, University of Verona, Verona, Italy
| |
Collapse
|
12
|
Nwabo Kamdje AH, Seke Etet PF, Simo RT, Vecchio L, Lukong KE, Krampera M. Emerging data supporting stromal cell therapeutic potential in cancer: reprogramming stromal cells of the tumor microenvironment for anti-cancer effects. Cancer Biol Med 2020; 17:828-841. [PMID: 33299638 PMCID: PMC7721102 DOI: 10.20892/j.issn.2095-3941.2020.0133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 08/20/2020] [Indexed: 02/03/2023] Open
Abstract
After more than a decade of controversy on the role of stromal cells in the tumor microenvironment, the emerging data shed light on pro-tumorigenic and potential anti-cancer factors, as well as on the roots of the discrepancies. We discuss the pro-tumorigenic effects of stromal cells, considering the effects of tumor drivers like hypoxia and tumor stiffness on these cells, as well as stromal cell-mediated adiposity and immunosuppression in the tumor microenvironment, and cancer initiating cells' cellular senescence and adaptive metabolism. We summarize the emerging data supporting stromal cell therapeutic potential in cancer, discuss the possibility to reprogram stromal cells of the tumor microenvironment for anti-cancer effects, and explore some causes of discrepancies on the roles of stromal cells in cancer in the available literature.
Collapse
Affiliation(s)
- Armel Hervé Nwabo Kamdje
- Department of Biomedical Sciences, University of Ngaoundere, Faculty of Science, Ngaoundere 454, Cameroon,Correspondence to: Armel Hervé Nwabo Kamdje, E-mail:
| | - Paul Faustin Seke Etet
- Department of Physiological Sciences and Biochemistry, University of Ngaoundéré, Garoua 454, Cameroon,Center for Sustainable Health and Development, Garoua 454, Cameroon
| | - Richard Tagne Simo
- Department of Biomedical Sciences, University of Ngaoundere, Faculty of Science, Ngaoundere 454, Cameroon
| | - Lorella Vecchio
- Center for Sustainable Health and Development, Garoua 454, Cameroon
| | - Kiven Erique Lukong
- Department of Biochemistry, Microbiology & Immunology, University of Saskatchewan, College of Medicine, Saskatoon SK S7N 5E5, Canada
| | - Mauro Krampera
- Department of Medicine, University of Verona, Section of Hematology, Stem Cell Research Laboratory, Verona 37134, Italy
| |
Collapse
|
13
|
Yang F, Li WB, Qu YW, Gao JX, Tang YS, Wang DJ, Pan YJ. Bone marrow mesenchymal stem cells induce M2 microglia polarization through PDGF-AA/MANF signaling. World J Stem Cells 2020; 12:633-658. [PMID: 32843919 PMCID: PMC7415242 DOI: 10.4252/wjsc.v12.i7.633] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 04/04/2020] [Accepted: 05/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cells (BMSCs) are capable of shifting the microglia/macrophages phenotype from M1 to M2, contributing to BMSCs-induced brain repair. However, the regulatory mechanism of BMSCs on microglia/macrophages after ischemic stroke is unclear. Recent evidence suggests that mesencephalic astrocyte–derived neurotrophic factor (MANF) and platelet-derived growth factor-AA (PDGF-AA)/MANF signaling regulate M1/M2 macrophage polarization.
AIM To investigate whether and how MANF or PDGF-AA/MANF signaling influences BMSCs-mediated M2 polarization.
METHODS We identified the secretion of MANF by BMSCs and developed transgenic BMSCs using a targeting small interfering RNA for knockdown of MANF expression. Using a rat middle cerebral artery occlusion (MCAO) model transplanted by BMSCs and BMSCs–microglia Transwell coculture system, the effect of BMSCs-induced downregulation of MANF expression on the phenotype of microglia/macrophages was tested by Western blot, quantitative reverse transcription-polymerase chain reaction, and immunofluorescence. Additionally, microglia were transfected with mimics of miR-30a*, which influenced expression of X-box binding protein (XBP) 1, a key transcription factor that synergized with activating transcription factor 6 (ATF6) to govern MANF expression. We examined the levels of miR-30a*, ATF6, XBP1, and MANF after PDGF-AA treatment in the activated microglia.
RESULTS Inhibition of MANF attenuated BMSCs-induced functional recovery and decreased M2 marker production, but increased M1 marker expression in vivo or in vitro. Furthermore, PDGF-AA treatment decreased miR-30a* expression, had no influence on the levels of ATF6, but enhanced expression of both XBP1 and MANF.
CONCLUSION BMSCs-mediated MANF paracrine signaling, in particular the PDGF-AA/miR-30a*/XBP1/MANF pathway, synergistically mediates BMSCs-induced M2 polarization.
Collapse
Affiliation(s)
- Fan Yang
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Wen-Bin Li
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Ye-Wei Qu
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Jin-Xing Gao
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yu-Shi Tang
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Dong-Jie Wang
- Department of Respiratory Medicine, The First Clinical College of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| | - Yu-Jun Pan
- Department of Neurology, The First Clinical College of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
14
|
Choe G, Kim SW, Park J, Park J, Kim S, Kim YS, Ahn Y, Jung DW, Williams DR, Lee JY. Anti-oxidant activity reinforced reduced graphene oxide/alginate microgels: Mesenchymal stem cell encapsulation and regeneration of infarcted hearts. Biomaterials 2019; 225:119513. [DOI: 10.1016/j.biomaterials.2019.119513] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/21/2022]
|
15
|
Gorabi AM, Bianconi V, Pirro M, Banach M, Sahebkar A. Regulation of cardiac stem cells by microRNAs: State-of-the-art. Biomed Pharmacother 2019; 120:109447. [PMID: 31580971 DOI: 10.1016/j.biopha.2019.109447] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/05/2019] [Accepted: 09/06/2019] [Indexed: 12/27/2022] Open
Abstract
Stem cells have a therapeutic potential in various medical conditions. In cases without sufficient response to conventional drug treatments, stem cells represent a next generation therapeutic strategy in cardiovascular diseases. Cardiac stem cells (CSCs), among a wide variety of stem cell sources, have been identified as a valid option for stem cell-based therapy in cardiovascular diseases. CSCs mainly act as a cell source to supply the physiological need for cardiovascular cells. However, they have been demonstrated to reproduce the myocardial cells under pathological settings. Despite their roles and functions have somewhat been clarified, molecular pathways underlying the regulatory mechanisms of CSCs are still not fully elucidated. Several studies have recently shown that different microRNAs (miRNAs) play a substantial role in regulating and controlling both the physiological and pathological proliferation and differentiation of stem cells. MiRNAs are small non-coding RNA molecules that regulate gene expression and may undergo aberrant expression levels during pathological conditions. Understanding the way through which miRNAs regulate CSC behavior may open up new horizons in modulating these cells in vitro to devise sophisticated approaches for treating patients with cardiovascular diseases. In this review article, we tried to discuss available evidence about the role of miRNAs in regulating CSCs.
Collapse
Affiliation(s)
- Armita Mahdavi Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Vanessa Bianconi
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Maciej Banach
- Department of Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Amirhossein Sahebkar
- Halal Research Center of IRI, FDA, Tehran, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
16
|
Liu Y, Zhao S, Luo L, Wang J, Zhu Z, Xiang Q, Deng Y, Zhao Z. Mesenchymal stem cell-derived exosomes ameliorate erection by reducing oxidative stress damage of corpus cavernosum in a rat model of artery injury. J Cell Mol Med 2019; 23:7462-7473. [PMID: 31512385 PMCID: PMC6815831 DOI: 10.1111/jcmm.14615] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/01/2019] [Accepted: 08/09/2019] [Indexed: 12/13/2022] Open
Abstract
Erectile dysfunction (ED) is a common ageing male's disease, and vascular ED accounts for the largest proportion of all types of ED. One of the mechanisms of vascular ED in the clinic is arterial insufficiency, which mainly caused by atherosclerosis, trauma and surgical. Moreover, oxidative stress damage after tissue ischemia usually aggravated the progress of ED. As a new way of acellular therapy, mesenchymal stem cell-derived exosomes (MSC-Exos) have great potential in ED treatment. In the current study, we have explored the mechanism of MSC-Exos therapy in a rat model of internal iliac artery injury-induced ED. Compared with intracavernous (IC) injection of phosphate-buffered saline after artery injury, of note, we observed that both mesenchymal stem cells (MSCs) and MSC-Exos through IC injection could improve the erectile function to varying degrees. More specifically, IC injection MSC-Exos could promote cavernous sinus endothelial formation, reduce the organization oxidative stress damage, and improve the nitric oxide synthase and smooth muscle content in the corpus cavernosum. With similar potency compared with the stem cell therapy and other unique advantages, IC injection of MSC- Exos could be an effective treatment to ameliorate erectile function in a rat model of arterial injury.
Collapse
Affiliation(s)
- Yangzhou Liu
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shankun Zhao
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.,Department of Urology, Zhejiang Taizhou Central Hospital (Taizhou University Hospital), Taizhou, China
| | - Lianmin Luo
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiamin Wang
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiguo Zhu
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qian Xiang
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yihan Deng
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhigang Zhao
- Department of Urology & Andrology, Minimally Invasive Surgery Center, Guangdong Provincial Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
17
|
Ng WH, Umar Fuaad MZ, Azmi SM, Leong YY, Yong YK, Ng AMH, Tan JJ. Guided evaluation and standardisation of mesenchymal stem cell culture conditions to generate conditioned medium favourable to cardiac c-kit cell growth. Cell Tissue Res 2018; 375:383-396. [PMID: 30232595 DOI: 10.1007/s00441-018-2918-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 09/03/2018] [Indexed: 12/20/2022]
Abstract
Mesenchymal stem cells (MSCs) are known to secrete cardioprotective paracrine factors that can potentially activate endogenous cardiac c-kit cells (CCs). This study aims to optimise MSC growth conditions and medium formulation for generating the conditioned medium (CdM) to facilitate CC growth and expansion in vitro. The quality of MSC-CdM after optimisation of seeding density during MSC stabilisation and medium formulation used during MSC stimulation including glucose, ascorbic acid, serum and oxygen levels and the effects of treatment concentration and repeated CdM harvesting were assessed based on CC viability in vitro under growth factor- and serum-deprived condition. Our data showed that functional CdM can be produced from MSCs with a density of 20,000 cells/cm2, which were stimulated using high glucose (25 mM), ascorbic acid supplemented, serum-free medium under normoxic condition. The generated CdM, when applied to growth factor- and serum-deprived medium at 1:1 ratio, improved CC viability, migration and proliferation in vitro. Such an effect could further be augmented by generating CdM concentrates without compromising CC gene and protein expressions, while retaining its capability to undergo differentiation to form endothelial, smooth muscle and cardiomyocytes. Nevertheless, CdM could not be repeatedly harvested from the same MSC culture, as the protein content and its effect on CC viability deteriorated after the first harvest. In conclusion, this study provides a proof-of-concept strategy to standardise the production of CdM from MSCs based on rapid, stepwise assessment of CC viability, thus enabling production of CdM favourable to CC growth for in vitro or clinical applications.
Collapse
Affiliation(s)
- Wai Hoe Ng
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Mimi Zulaikha Umar Fuaad
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Siti Maisura Azmi
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Yin Yee Leong
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Angela Min Hwei Ng
- Tissue Engineering Centre, Universiti Kebangsaan Malaysia Medical Centre, 56000, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur, Malaysia
| | - Jun Jie Tan
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, 13200, Kepala Batas, Penang, Malaysia.
| |
Collapse
|
18
|
Fahy N, Alini M, Stoddart MJ. Mechanical stimulation of mesenchymal stem cells: Implications for cartilage tissue engineering. J Orthop Res 2018; 36:52-63. [PMID: 28763118 DOI: 10.1002/jor.23670] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/24/2017] [Indexed: 02/04/2023]
Abstract
Articular cartilage is a load-bearing tissue playing a crucial mechanical role in diarthrodial joints, facilitating joint articulation, and minimizing wear. The significance of biomechanical stimuli in the development of cartilage and maintenance of chondrocyte phenotype in adult tissues has been well documented. Furthermore, dysregulated loading is associated with cartilage pathology highlighting the importance of mechanical cues in cartilage homeostasis. The repair of damaged articular cartilage resulting from trauma or degenerative joint disease poses a major challenge due to a low intrinsic capacity of cartilage for self-renewal, attributable to its avascular nature. Bone marrow-derived mesenchymal stem cells (MSCs) are considered a promising cell type for cartilage replacement strategies due to their chondrogenic differentiation potential. Chondrogenesis of MSCs is influenced not only by biological factors but also by the environment itself, and various efforts to date have focused on harnessing biomechanics to enhance chondrogenic differentiation of MSCs. Furthermore, recapitulating mechanical cues associated with cartilage development and homeostasis in vivo, may facilitate the development of a cellular phenotype resembling native articular cartilage. The goal of this review is to summarize current literature examining the effect of mechanical cues on cartilage homeostasis, disease, and MSC chondrogenesis. The role of biological factors produced by MSCs in response to mechanical loading will also be examined. An in-depth understanding of the impact of mechanical stimulation on the chondrogenic differentiation of MSCs in terms of endogenous bioactive factor production and signaling pathways involved, may identify therapeutic targets and facilitate the development of more robust strategies for cartilage replacement using MSCs. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:52-63, 2018.
Collapse
Affiliation(s)
- Niamh Fahy
- AO Research Institute Davos, Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | |
Collapse
|
19
|
Nwabo Kamdje AH, Kamga PT, Simo RT, Vecchio L, Seke Etet PF, Muller JM, Bassi G, Lukong E, Goel RK, Amvene JM, Krampera M. Mesenchymal stromal cells' role in tumor microenvironment: involvement of signaling pathways. Cancer Biol Med 2017; 14:129-141. [PMID: 28607804 PMCID: PMC5444925 DOI: 10.20892/j.issn.2095-3941.2016.0033] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are adult multipotent stem cells residing as pericytes in various tissues and organs where they can differentiate into specialized cells to replace dying cells and damaged tissues. These cells are commonly found at injury sites and in tumors that are known to behave like " wounds that do not heal." In this article, we discuss the mechanisms of MSCs in migrating, homing, and repairing injured tissues. We also review a number of reports showing that tumor microenvironment triggers plasticity mechanisms in MSCs to induce malignant neoplastic tissue formation, maintenance, and chemoresistance, as well as tumor growth. The antitumor properties and therapeutic potential of MSCs are also discussed.
Collapse
Affiliation(s)
| | - Paul Takam Kamga
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Richard Tagne Simo
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Lorella Vecchio
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | | | - Jean Marc Muller
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Giulio Bassi
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Erique Lukong
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Raghuveera Kumar Goel
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Jeremie Mbo Amvene
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| | - Mauro Krampera
- Department of Biomedical Sciences, University of Ngaoundere, Ngaoundere 454, Cameroon
| |
Collapse
|
20
|
Yan T, Venkat P, Chopp M, Zacharek A, Ning R, Roberts C, Zhang Y, Lu M, Chen J. Neurorestorative Responses to Delayed Human Mesenchymal Stromal Cells Treatment of Stroke in Type 2 Diabetic Rats. Stroke 2016; 47:2850-2858. [PMID: 27729575 DOI: 10.1161/strokeaha.116.014686] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/14/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND PURPOSE Comorbidity of diabetes mellitus and stroke results in worse functional outcome, poor long-term recovery, and extensive vascular damage. We investigated the neurorestorative effects and mechanisms of stroke treatment with human bone marrow-derived mesenchymal stromal cells (hMSCs) in type 2 diabetes mellitus (T2DM) rats. METHODS Adult male Wistar rats were induced with T2DM, subjected to 2 hours of middle cerebral artery occlusion (MCAo) and treated via tail-vein injection with (1) PBS (n=8) and (2) hMSCs (n=10; 5×106) at 3 days after MCAo. RESULTS In T2DM rats, hMSCs administered at 3 days after MCAo significantly improves neurological function without affecting blood glucose, infarct volume, and incidence of brain hemorrhage in comparison to T2DM-MCAo PBS-treated rats. Delayed hMSC treatment of T2DM stroke significantly improves blood-brain barrier integrity, increases vascular and arterial density and cerebral vascular perfusion, and promotes neuroblast cell migration and white matter remodeling as indicated by increased doublecortin, axon, myelin, and neurofilament density, respectively. Delayed hMSC treatment significantly increases platelet-derived growth factor expression in the ischemic brain, decreases proinflammatory M1 macrophage and increases anti-inflammatory M2 macrophage compared to PBS-treated T2DM-MCAo rats. In vitro data show that hMSCs increase subventricular zone explant cell migration and primary cortical neuron neurite outgrowth, whereas inhibition of platelet-derived growth factor decreases hMSC-induced subventricular zone cell migration and axonal outgrowth. CONCLUSIONS In T2DM stroke rats, delayed hMSC treatment significantly improves neurological functional outcome and increases neurorestorative effects and M2 macrophage polarization. Increasing brain platelet-derived growth factor expression may contribute to hMSC-induced neurorestoration.
Collapse
Affiliation(s)
- Tao Yan
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Poornima Venkat
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Michael Chopp
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Alex Zacharek
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Ruizhuo Ning
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Cynthia Roberts
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Yi Zhang
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Mei Lu
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Jieli Chen
- From the Tianjin Neurological and Gerontology Institute, Department of Neurology of Tianjin Medical University General Hospital, China (T.Y., J.C.); Department of Neurology (T.Y., P.V., M.C., A.Z., R.N., C.R., Y.Z., J.C.) and Department of Biostatistics and Research Epidemiology (M.L.), Henry Ford Hospital, Detroit, MI; and Department of Physics, Oakland University, Rochester, MI (M.C.).
| |
Collapse
|
21
|
Tao L, Bei Y, Zhang H, Xiao J, Li X. Exercise for the heart: signaling pathways. Oncotarget 2016; 6:20773-84. [PMID: 26318584 PMCID: PMC4673228 DOI: 10.18632/oncotarget.4770] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 07/10/2015] [Indexed: 12/30/2022] Open
Abstract
Physical exercise, a potent functional intervention in protecting against cardiovascular diseases, is a hot topic in recent years. Exercise has been shown to reduce cardiac risk factors, protect against myocardial damage, and increase cardiac function. This improves quality of life and decreases mortality and morbidity in a variety of cardiovascular diseases, including myocardial infarction, cardiac ischemia/reperfusion injury, diabetic cardiomyopathy, cardiac aging, and pulmonary hypertension. The cellular adaptation to exercise can be associated with both endogenous and exogenous factors: (1) exercise induces cardiac growth via hypertrophy and renewal of cardiomyocytes, and (2) exercise induces endothelial progenitor cells to proliferate, migrate and differentiate into mature endothelial cells, giving rise to endothelial regeneration and angiogenesis. The cellular adaptations associated with exercise are due to the activation of several signaling pathways, in particular, the growth factor neuregulin1 (NRG1)-ErbB4-C/EBPβ and insulin-like growth factor (IGF)-1-PI3k-Akt signaling pathways. Of interest, microRNAs (miRNAs, miRs) such as miR-222 also play a major role in the beneficial effects of exercise. Thus, exploring the mechanisms mediating exercise-induced benefits will be instrumental for devising new effective therapies against cardiovascular diseases.
Collapse
Affiliation(s)
- Lichan Tao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yihua Bei
- Regeneration and Ageing Lab and Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai 200444, China.,Shanghai Key Laboratory of Bio-Energy Crops, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Haifeng Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Junjie Xiao
- Regeneration and Ageing Lab and Experimental Center of Life Sciences, School of Life Science, Shanghai University, Shanghai 200444, China.,Shanghai Key Laboratory of Bio-Energy Crops, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xinli Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
22
|
Karantalis V, Suncion-Loescher VY, Bagno L, Golpanian S, Wolf A, Sanina C, Premer C, Kanelidis AJ, McCall F, Wang B, Balkan W, Rodriguez J, Rosado M, Morales A, Hatzistergos K, Natsumeda M, Margitich I, Schulman IH, Gomes SA, Mushtaq M, DiFede DL, Fishman JE, Pattany P, Zambrano JP, Heldman AW, Hare JM. Synergistic Effects of Combined Cell Therapy for Chronic Ischemic Cardiomyopathy. J Am Coll Cardiol 2016; 66:1990-1999. [PMID: 26516002 DOI: 10.1016/j.jacc.2015.08.879] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 08/12/2015] [Accepted: 08/17/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Both bone marrow-derived mesenchymal stem cells (MSCs) and c-kit(+) cardiac stem cells (CSCs) improve left ventricular remodeling in porcine models and clinical trials. Using xenogeneic (human) cells in immunosuppressed animals with acute ischemic heart disease, we previously showed that these 2 cell types act synergistically. OBJECTIVES To more accurately model clinical applications for heart failure, this study tested whether the combination of autologous MSCs and CSCs produce greater improvement in cardiac performance than MSCs alone in a nonimmunosuppressed porcine model of chronic ischemic cardiomyopathy. METHODS Three months after ischemia/reperfusion injury, Göttingen swine received transendocardial injections with MSCs alone (n = 6) or in combination with cardiac-derived CSCs (n = 8), or placebo (vehicle; n = 6). Cardiac functional and anatomic parameters were assessed using cardiac magnetic resonance at baseline and before and after therapy. RESULTS Both groups of cell-treated animals exhibited significantly reduced scar size (MSCs -44.1 ± 6.8%; CSC/MSC -37.2 ± 5.4%; placebo -12.9 ± 4.2%; p < 0.0001), increased viable tissue, and improved wall motion relative to placebo 3 months post-injection. Ejection fraction (EF) improved (MSCs 2.9 ± 1.6 EF units; CSC/MSC 6.9 ± 2.8 EF units; placebo 2.5 ± 1.6 EF units; p = 0.0009), as did stroke volume, cardiac output, and diastolic strain only in the combination-treated animals, which also exhibited increased cardiomyocyte mitotic activity. CONCLUSIONS These findings illustrate that interactions between MSCs and CSCs enhance cardiac performance more than MSCs alone, establish the safety of autologous cell combination strategies, and support the development of second-generation cell therapeutic products.
Collapse
Affiliation(s)
- Vasileios Karantalis
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Viky Y Suncion-Loescher
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Luiza Bagno
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Samuel Golpanian
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Ariel Wolf
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Cristina Sanina
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Courtney Premer
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Anthony J Kanelidis
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Frederic McCall
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Bo Wang
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Wayne Balkan
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Jose Rodriguez
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Marcos Rosado
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Azorides Morales
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida
| | - Konstantinos Hatzistergos
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Makoto Natsumeda
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Irene Margitich
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Ivonne Hernandez Schulman
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Samirah A Gomes
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Muzammil Mushtaq
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Darcy L DiFede
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Joel E Fishman
- Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | - Pradip Pattany
- Department of Radiology, University of Miami Miller School of Medicine, Miami, Florida
| | | | - Alan W Heldman
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida
| | - Joshua M Hare
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
23
|
Yang CJ, Yang J, Yang J, Fan ZX. Cardiac atrial appendage stem cells therapy: a novel and promising approach for myocardial reparation after MI. Int J Cardiol 2016; 203:1153-1154. [PMID: 26478524 DOI: 10.1016/j.ijcard.2015.10.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 10/04/2015] [Indexed: 11/23/2022]
Affiliation(s)
- Chao-Jun Yang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000 Hubei Province, China
| | - Jun Yang
- Department of Cardiology, the First College of Clinical Medical Sciences, China Three Gorges University, Yichang 443000 Hubei Province, China.
| | - Jian Yang
- Department of Cardiology, the First College of Clinical Medical Sciences, China Three Gorges University, Yichang 443000 Hubei Province, China
| | - Zhi-Xing Fan
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000 Hubei Province, China; Department of Cardiology, the First College of Clinical Medical Sciences, China Three Gorges University, Yichang 443000 Hubei Province, China
| |
Collapse
|
24
|
BMSCs Interactions with Adventitial Fibroblasts Display Smooth Muscle Cell Lineage Potential in Differentiation and Migration That Contributes to Neointimal Formation. Stem Cells Int 2016; 2016:3196071. [PMID: 26880952 PMCID: PMC4736561 DOI: 10.1155/2016/3196071] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/04/2015] [Accepted: 11/05/2015] [Indexed: 12/30/2022] Open
Abstract
In this study a model of simulated vascular injury in vitro was used to study the characterization of bone-marrow-derived mesenchymal stem cells (BMSCs) morphology and to investigate the differentiation and migration of BMSCs in the presence of adventitial fibroblasts. BMSCs from rats were indirectly cocultured with adventitial fibroblasts in a transwell chamber apparatus for 7 days, and clonogenic assays demonstrated that BMSCs could be differentiated into smooth muscle-like cells with this process, including smooth muscle α-actin (α-SMA) expression by immunofluorescence staining. Cell morphology of BMSCs was assessed by inverted microscope, while cell proliferation was assessed by MTT assay. The expressions of TGF-β1, MMP-1, and NF-κB were detected by immunofluorescence staining and Smad3 mRNA was measured by reverse transcription PCR. Migration ability of BMSCs with DAPI-labeled nuclei was measured by laser confocal microscopy. Our results demonstrate that indirect interactions with adventitial fibroblasts can induce proliferation, differentiation, and migration of BMSCs that can actively participate in neointimal formation. Our results indicate that the pathogenesis of vascular remodeling might perform via TGF-β1/Smad3 signal transduction pathways.
Collapse
|
25
|
Fanton Y, Robic B, Rummens JL, Daniëls A, Windmolders S, Willems L, Jamaer L, Dubois J, Bijnens E, Heuts N, Notelaers K, Paesen R, Ameloot M, Mees U, Bito V, Declercq J, Hensen K, Koninckx R, Hendrikx M. Possibilities and limitations for co-transplantation of cardiac atrial appendage stem cells and mesenchymal stem cells for myocardial repair. Int J Cardiol 2015; 203:1155-6. [PMID: 26549562 DOI: 10.1016/j.ijcard.2015.10.141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 10/18/2015] [Indexed: 11/19/2022]
Affiliation(s)
- Yanick Fanton
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium.
| | - Boris Robic
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Cardiothoracic Surgery, Jessa Hospital, Hasselt, Belgium
| | - Jean-Luc Rummens
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Annick Daniëls
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium
| | - Severina Windmolders
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Leen Willems
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Luc Jamaer
- Department of Cardiac Anesthesia, Jessa Hospital, Hasselt, Belgium
| | - Jasperina Dubois
- Department of Cardiac Anesthesia, Jessa Hospital, Hasselt, Belgium
| | - Eric Bijnens
- MRI Unit-Department of Radiology, Jessa Hospital, Hasselt, Belgium
| | - Nic Heuts
- MRI Unit-Department of Radiology, Jessa Hospital, Hasselt, Belgium
| | - Kristof Notelaers
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Rik Paesen
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Marcel Ameloot
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Urbain Mees
- Department of Cardiothoracic Surgery, Jessa Hospital, Hasselt, Belgium
| | - Virginie Bito
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| | - Jeroen Declercq
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Karen Hensen
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Remco Koninckx
- Laboratory of Experimental Hematology, Jessa Hospital, Hasselt, Belgium; Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Marc Hendrikx
- Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Cardiothoracic Surgery, Jessa Hospital, Hasselt, Belgium
| |
Collapse
|
26
|
Ho YS, Tsai WH, Lin FC, Huang WP, Lin LC, Wu SM, Liu YR, Chen WP. Cardioprotective Actions of TGFβRI Inhibition Through Stimulating Autocrine/Paracrine of Survivin and Inhibiting Wnt in Cardiac Progenitors. Stem Cells 2015; 34:445-55. [PMID: 26418219 DOI: 10.1002/stem.2216] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/17/2015] [Accepted: 09/14/2015] [Indexed: 01/10/2023]
Abstract
Heart failure due to myocardial infarction (MI) is a major cause of morbidity and mortality in the world. We found previously that A83-01, a TGFβRI inhibitor, could facilitate cardiac repair in post-MI mice and induce the expansion of a Nkx2.5 + cardiomyoblast population. This study aimed to investigate the key autocrine/paracrine factors regulated by A83-01 in the injured heart and the mechanism of cardioprotection by this molecule. Using a previously described transgenic Nkx2.5 enhancer-green fluorescent protein (GFP) reporter mice, we isolated cardiac progenitor cells (CPC) including Nkx2.5-GFP + (Nkx2.5+), sca1+, and Nkx2.5+/sca1 + cells. A83-01 was found to induce proliferation of these three subpopulations mainly through increasing Birc5 expression in the MEK/ERK-dependent pathway. Survivin, encoded by Birc5, could also directly proliferate Nkx2.5 + cells and enhance cultured cardiomyocytes viability. A83-01 could also reverse the downregulation of Birc5 in postinjured mice hearts (n = 6) to expand CPCs. Moreover, the increased Wnt3a in postinjured hearts could decrease CPCs, which could be reversed by A83-01 via inhibiting Fzd6 and Wnt1-induced signaling protein 1 expressions in CPCs. Next, we used inducible αMHC-cre/mTmG mice to label cardiomyocytes with GFP and nonmyocytes with RFP. We found A83-01 preserved more GFP + myocytes (68.6% ± 3.1% vs. 80.9% ± 3.0%; p < .05, n = 6) and fewer renewed RFP + myocytes (0.026% ± 0.005% vs. 0.062% ± 0.008%; p < .05, n = 6) in parallel with less cardiac fibrosis in isoprenaline-injected mice treated with A83-01. TGFβRI inhibition in an injured adult heart could both stimulate the autocrine/paracrine activity of survivin and inhibit Wnt in CPCs to mediate cardioprotection and improve cardiac function.
Collapse
Affiliation(s)
- Yu-Sian Ho
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Wan-Hsuan Tsai
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Fen-Chiung Lin
- Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei City, Taiwan.,Division of Cardiology, Linkou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Wei-Pang Huang
- Department of Life Science, National Taiwan University, Taipei City, Taiwan
| | - Lung-Chun Lin
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Sean M Wu
- Division of Cardiovascular Medicine, Department of Medicine, Cardiovascular Institute, Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Yu-Ru Liu
- Laboratory Animal Center, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Wen-Pin Chen
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei City, Taiwan
| |
Collapse
|
27
|
Yao Y, Huang J, Geng Y, Qian H, Wang F, Liu X, Shang M, Nie S, Liu N, Du X, Dong J, Ma C. Paracrine action of mesenchymal stem cells revealed by single cell gene profiling in infarcted murine hearts. PLoS One 2015; 10:e0129164. [PMID: 26043119 PMCID: PMC4456391 DOI: 10.1371/journal.pone.0129164] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 05/05/2015] [Indexed: 11/30/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have been recently demonstrated as a promising stem cell type to rescue damaged myocardium after acute infarction. One of the most important mechanisms underlying their therapeutic effects is the secretion of paracrine factors. However, the expression profile of paracrine factors of MSCs in infarcted hearts, especially at single cell level, is poorly defined. Methods and Results We aimed to depict the transcriptional profile of paracrine factors secreted by MSCs in vivo, with particular interest in the comparison between normal and infarcted hearts. Bone marrow mesenchymal stem cells were isolated and injected into mice hearts immediately after infarction surgery. Bioluminescence imaging (BLI) indicated a proportion of cells still alive even up to 10 days post surgery. Paralleled with survived cells, cardiac function was significantly improved after MSC injection compared to that in PBS-injected mice, indicated by MRI and histology. Despite increased number of vessels in MSC-injected hearts, endothelial cells and cardiomyocytes transdifferentiation were not observed in infarcted hearts 5 days after infarction. Furthermore, laser capture microdissection (LCM) followed by high through-put real time PCR was employed in our study, uncovering that the injected MSCs, compared to local cardiomyocytes, displayed elevated levels of secreted factors. To further investigate the regulation of those factors, we performed single cell analysis to dissect the gene expression profile of MSCs at single cell level in infarcted and normal hearts, respectively. Consistent with the in vivo observation, a similar regulation pattern of those factors was detected in cultured MSCs under hypoxia. Conclusions Our study, for the first time, elucidated gene expression profiles, as well as regulation of paracrine factors, of MSCs at single cell level in vivo, indicating that paracrine factors from MSCs account for the improvement of cardiac function after infarction.
Collapse
Affiliation(s)
- Yan Yao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, P. R. China
- * E-mail:
| | - Ji Huang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, P. R. China
| | - Yongjian Geng
- Center for Cardiovascular Biology and Atherosclerosis, Department of Internal Medicine, The University of Texas, Health Science Center at Houston, Medical School, Texas Heart Institute, Houston, TX, United States of America
| | - Haiyan Qian
- Center for Coronary Heart Disease, Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Fan Wang
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Xiaohui Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, P. R. China
| | - Meisheng Shang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, P. R. China
| | - Shaoping Nie
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, P. R. China
| | - Nian Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, P. R. China
| | - Xin Du
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, P. R. China
| | - Jianzeng Dong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, P. R. China
| | - Changsheng Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, P. R. China
| |
Collapse
|
28
|
Noseda M, Harada M, McSweeney S, Leja T, Belian E, Stuckey DJ, Abreu Paiva MS, Habib J, Macaulay I, de Smith AJ, al-Beidh F, Sampson R, Lumbers RT, Rao P, Harding SE, Blakemore AIF, Eirik Jacobsen S, Barahona M, Schneider MD. PDGFRα demarcates the cardiogenic clonogenic Sca1+ stem/progenitor cell in adult murine myocardium. Nat Commun 2015; 6:6930. [PMID: 25980517 PMCID: PMC4479024 DOI: 10.1038/ncomms7930] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 03/16/2015] [Indexed: 12/24/2022] Open
Abstract
Cardiac progenitor/stem cells in adult hearts represent an attractive therapeutic target for heart regeneration, though (inter)-relationships among reported cells remain obscure. Using single-cell qRT-PCR and clonal analyses, here we define four subpopulations of cardiac progenitor/stem cells in adult mouse myocardium all sharing stem cell antigen-1 (Sca1), based on side population (SP) phenotype, PECAM-1 (CD31) and platelet-derived growth factor receptor-α (PDGFRα) expression. SP status predicts clonogenicity and cardiogenic gene expression (Gata4/6, Hand2 and Tbx5/20), properties segregating more specifically to PDGFRα(+) cells. Clonal progeny of single Sca1(+) SP cells show cardiomyocyte, endothelial and smooth muscle lineage potential after cardiac grafting, augmenting cardiac function although durable engraftment is rare. PDGFRα(-) cells are characterized by Kdr/Flk1, Cdh5, CD31 and lack of clonogenicity. PDGFRα(+)/CD31(-) cells derive from cells formerly expressing Mesp1, Nkx2-5, Isl1, Gata5 and Wt1, distinct from PDGFRα(-)/CD31(+) cells (Gata5 low; Flk1 and Tie2 high). Thus, PDGFRα demarcates the clonogenic cardiogenic Sca1(+) stem/progenitor cell.
Collapse
Affiliation(s)
- Michela Noseda
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Mutsuo Harada
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Sara McSweeney
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Thomas Leja
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Elisa Belian
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Daniel J. Stuckey
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
- Centre for Advanced Biomedical Imaging (CABI), University College London, London WC1E 6DD, UK
| | - Marta S. Abreu Paiva
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Josef Habib
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
- Department of Biomedical Engineering, King's College London, London SE1 7EH, UK
| | - Iain Macaulay
- Haematopoietic Stem Cell Biology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Adam J. de Smith
- Department of Medicine, Imperial College London, London SW7 2AZ, UK
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California 94143, USA
| | - Farah al-Beidh
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Robert Sampson
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - R. Thomas Lumbers
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | - Pulivarthi Rao
- Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Sian E. Harding
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| | | | - Sten Eirik Jacobsen
- Haematopoietic Stem Cell Biology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Mauricio Barahona
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- Department of Mathematics, Imperial College London, London SW7 2AZ, UK
| | - Michael D. Schneider
- British Heart Foundation Centre of Research Excellence, Imperial College London, London SW7 2AZ, UK
- National Heart and Lung Institute, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
29
|
Bara JJ, Turner S, Roberts S, Griffiths G, Benson R, Trivedi JM, Wright KT. High content and high throughput screening to assess the angiogenic and neurogenic actions of mesenchymal stem cells in vitro. Exp Cell Res 2015; 333:93-104. [DOI: 10.1016/j.yexcr.2014.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 12/16/2014] [Accepted: 12/30/2014] [Indexed: 01/01/2023]
|
30
|
Murata D, Tokunaga S, Tamura T, Kawaguchi H, Miyoshi N, Fujiki M, Nakayama K, Misumi K. A preliminary study of osteochondral regeneration using a scaffold-free three-dimensional construct of porcine adipose tissue-derived mesenchymal stem cells. J Orthop Surg Res 2015; 10:35. [PMID: 25890366 PMCID: PMC4389925 DOI: 10.1186/s13018-015-0173-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 02/25/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a major joint disease in humans and many other animals. Consequently, medical countermeasures for OA have been investigated diligently. This study was designed to examine the regeneration of articular cartilage and subchondral bone using three-dimensional (3D) constructs of adipose tissue-derived mesenchymal stem cells (AT-MSCs). METHODS AT-MSCs were isolated and expanded until required for genetical and immunological analysis and construct creation. A construct consisting of about 760 spheroids that each contained 5.0 × 10(4) autologous AT-MSCs was implanted into an osteochondral defect (diameter: 4 mm; depth: 6 mm) created in the femoral trochlear groove of two adult microminipigs. After implantation, the defects were monitored by computed tomography every month for 6 months in animal no. 1 and 12 months in animal no. 2. RESULTS AT-MSCs were confirmed to express the premature genes and to be positive for CD90 and CD105 and negative for CD34 and CD45. Under specific nutrient conditions, the AT-MSCs differentiated into osteogenic, chondrogenic, and adipogenic lineages, as evidenced by the expressions of related marker genes and the production of appropriate matrix molecules. A radiopaque area emerged from the boundary between the bone and the implant and increased more steadily upward and inward for the implants in both animal no. 1 and animal no. 2. The histopathology of the implants after 6 months revealed active endochondral ossification underneath the plump fibrocartilage in animal no. 1. The histopathology after 12 months in animal no. 2 showed not only that the diminishing fibrocartilage was as thick as the surrounding normal cartilage but also that massive subchondral bone was present. CONCLUSIONS The present results suggest that implantation of a scaffold-free 3D construct of AT-MSCs into an osteochondral defect may induce regeneration of the original structure of the cartilage and subchondral bone over the course of 1 year, although more experimental cases are needed.
Collapse
Affiliation(s)
- Daiki Murata
- Veterinary Surgery, Department of Veterinary Clinical Science, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan.
| | - Satoshi Tokunaga
- Veterinary Teaching Hospital, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan.
| | - Tadashi Tamura
- Cyfuse Biomedical K.K., 1-1 Maidashi 3-chome, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Hiroaki Kawaguchi
- Veterinary Pathology, Department of Pathological and Preventive Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan.
| | - Noriaki Miyoshi
- Veterinary Pathology, Department of Pathological and Preventive Sciences, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan.
| | - Makoto Fujiki
- Veterinary Surgery, Department of Veterinary Clinical Science, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan.
| | - Koichi Nakayama
- Department of Advanced Technology Fusion, Graduate School of Science and Engineering, Saga University, Honjyo 1-chome, Honjyo-cho, Saga, 840-8502, Japan.
| | - Kazuhiro Misumi
- Veterinary Surgery, Department of Veterinary Clinical Science, Joint Faculty of Veterinary Medicine, Kagoshima University, 21-24 Korimoto 1-chome, Kagoshima, 890-0065, Japan.
| |
Collapse
|
31
|
Gautam M, Fujita D, Kimura K, Ichikawa H, Izawa A, Hirose M, Kashihara T, Yamada M, Takahashi M, Ikeda U, Shiba Y. Transplantation of adipose tissue-derived stem cells improves cardiac contractile function and electrical stability in a rat myocardial infarction model. J Mol Cell Cardiol 2015; 81:139-49. [PMID: 25724725 DOI: 10.1016/j.yjmcc.2015.02.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 01/08/2023]
Abstract
The transplantation of adipose tissue-derived stem cells (ADSCs) improves cardiac contractility after myocardial infarction (MI); however, little is known about the electrophysiological consequences of transplantation. The purpose of this study was to clarify whether the transplantation of ADSCs increases or decreases the incidence of ventricular tachyarrhythmias (VT) in a rat model of MI. MI was induced experimentally by permanent occlusion of the left anterior descending artery of Lewis rats. ADSCs were harvested from GFP-transgenic rats, and were cultured until passage four. ADSCs (10×10(6)) resuspended in 100μL saline or pro-survival cocktail (PSC), which enhances cardiac graft survival, were injected directly into syngeneic rat hearts 1week after MI. The recipients of ADSCs suspended in PSC had a larger graft area compared with those receiving ASDCs suspended in saline at 1week post-transplantation (number of graft cells/section: 148.7±10.6 vs. 22.4±3.4, p<0.05, n=5/group). Thereafter, all ADSC recipients were transplanted with ASDCs in PSC. ADSCs were transplanted into infarcted hearts, and the mechanical and electrophysiological functions were assessed. Echocardiography revealed that ADSC recipients had improved contractile function compared with those receiving PSC vehicle (fractional shortening: 21.1±0.9 vs. 14.1±1.2, p<0.05, n≥12/group). Four weeks post-transplantation, VT was induced via in vivo programmed electrical stimulation. The recipients of ADSCs showed a significantly lower incidence of induced VT compared with the control (31.3% vs. 83.3%, p<0.05, n≥12/group). To understand the electrical activity following transplantation, we performed ex vivo optical mapping using a voltage sensitive dye, and found that ADSC transplantation decreased conduction velocity and its dispersion in the peri-infarct area. These results suggest that ADSC transplantation improved cardiac mechanical and electrophysiological functions in subacute MI.
Collapse
Affiliation(s)
- Milan Gautam
- Department of Cardiovascular Medicine, Shinshu University, Matsumoto, Japan
| | - Daiki Fujita
- Department of Anatomy and Organ Technology, Shinshu University, Matsumoto, Japan; Department of Biotechnology and Biomedical Engineering, Institute for Biomedical Sciences, Shinshu University, Matsumoto, Japan
| | - Kazuhiro Kimura
- Department of Cardiovascular Medicine, Shinshu University, Matsumoto, Japan
| | - Hinako Ichikawa
- Department of Cardiovascular Medicine, Shinshu University, Matsumoto, Japan
| | - Atsushi Izawa
- Department of Cardiovascular Medicine, Shinshu University, Matsumoto, Japan
| | - Masamichi Hirose
- Department of Molecular and Cellular Pharmacology, Iwate Medical University, Iwate, Japan
| | | | | | - Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Uichi Ikeda
- Department of Cardiovascular Medicine, Shinshu University, Matsumoto, Japan
| | - Yuji Shiba
- Department of Cardiovascular Medicine, Shinshu University, Matsumoto, Japan; Department of Biotechnology and Biomedical Engineering, Institute for Biomedical Sciences, Shinshu University, Matsumoto, Japan.
| |
Collapse
|
32
|
The Role of MicroRNAs in Cardiac Stem Cells. Stem Cells Int 2015; 2015:194894. [PMID: 25802528 PMCID: PMC4329769 DOI: 10.1155/2015/194894] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/14/2014] [Accepted: 01/05/2015] [Indexed: 12/12/2022] Open
Abstract
Stem cells are considered as the next generation drug treatment in patients with cardiovascular disease who are resistant to conventional treatment. Among several stem cells used in the clinical setting, cardiac stem cells (CSCs) which reside in the myocardium and epicardium of the heart have been shown to be an effective option for the source of stem cells. In normal circumstances, CSCs primarily function as a cell store to replace the physiologically depleted cardiovascular cells, while under the diseased condition they have been shown to experimentally regenerate the diseased myocardium. In spite of their major functional role, molecular mechanisms regulating the CSCs proliferation and differentiation are still unknown. MicroRNAs (miRs) are small, noncoding RNA molecules that regulate gene expression at the posttranscriptional level. Recent studies have demonstrated the important role of miRs in regulating stem cell proliferation and differentiation, as well as other physiological and pathological processes related to stem cell function. This review summarises the current understanding of the role of miRs in CSCs. A deeper understanding of the mechanisms by which miRs regulate CSCs may lead to advances in the mode of stem cell therapies for the treatment of cardiovascular diseases.
Collapse
|
33
|
Bussche L, Van de Walle GR. Peripheral Blood-Derived Mesenchymal Stromal Cells Promote Angiogenesis via Paracrine Stimulation of Vascular Endothelial Growth Factor Secretion in the Equine Model. Stem Cells Transl Med 2014; 3:1514-25. [PMID: 25313202 DOI: 10.5966/sctm.2014-0138] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have received much attention as a potential treatment of ischemic diseases, including ischemic tissue injury and cardiac failure. The beneficial effects of MSCs are thought to be mediated by their ability to provide proangiogenic factors, creating a favorable microenvironment that results in neovascularization and tissue regeneration. To study this in more detail and to explore the potential of the horse as a valuable translational model, the objectives of the present study were to examine the presence of angiogenic stimulating factors in the conditioned medium (CM) of peripheral blood-derived equine mesenchymal stromal cells (PB-MSCs) and to study their in vitro effect on angiogenesis-related endothelial cell (EC) behavior, including proliferation and vessel formation. Our salient findings were that CM from PB-MSCs contained significant levels of several proangiogenic factors. Furthermore, we found that CM could induce angiogenesis in equine vascular ECs and confirmed that endothelin-1, insulin growth factor binding protein 2, interleukin-8, and platelet-derived growth factor-AA, but not urokinase-type plasminogen activator, were responsible for this enhanced EC network formation by increasing the expression level of vascular endothelial growth factor-A, an important angiogenesis stimulator.
Collapse
Affiliation(s)
- Leen Bussche
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
34
|
Pankajakshan D, Agrawal DK. Mesenchymal Stem Cell Paracrine Factors in Vascular Repair and Regeneration. ACTA ACUST UNITED AC 2014; 1. [PMID: 28890954 DOI: 10.19104/jbtr.2014.107] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mesenchymal stem cell therapy show great optimism in the treatment of several diseases. MSCs are attractive candidates for cell therapy because of easy isolation, high expansion potential giving unlimited pool of transplantable cells, low immunogenicity, amenability to ex vivo genetic modification, and multipotency. The stem cells orchestrate the repair process by various mechanisms such as transdifferentiation, cell fusion, microvesicles or exosomes and most importantly by secreting paracrine factors. The MSCs release several angiogenic, mitogenic, anti-apoptotic, anti-inflammatory and anti-oxidative factors that play fundamental role in regulating tissue repair in various vascular and cardiac diseases. The therapeutic release of these factors by the cells can be enhanced by several strategies like genetic modification, physiological and pharmacological preconditioning, improved cell culture and selection methods, and biomaterial based approaches. The current review describes the impact of paracrine factors released by MSCs on vascular repair and regeneration in myocardial infarction, restenosis and peripheral artery disease, and the various strategies adopted to enhance the release of these paracrine factors to enhance organ function.
Collapse
Affiliation(s)
- Divya Pankajakshan
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| | - Devendra K Agrawal
- Department of Biomedical Sciences, Creighton University School of Medicine, Omaha, NE, USA
| |
Collapse
|