1
|
Lee EJ, Jang WB, Choi J, Lim HJ, Park S, Rethineswaran VK, Ha JS, Yun J, Hong YJ, Choi YJ, Kwon SM. The Protective Role of Glutathione against Doxorubicin-Induced Cardiotoxicity in Human Cardiac Progenitor Cells. Int J Mol Sci 2023; 24:12070. [PMID: 37569446 PMCID: PMC10419046 DOI: 10.3390/ijms241512070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/22/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
This study investigated the protective effect of glutathione (GSH), an antioxidant drug, against doxorubicin (DOX)-induced cardiotoxicity. Human cardiac progenitor cells (hCPCs) treated with DOX (250 to 500 nM) showed increased viability and reduced ROS generation and apoptosis with GSH treatment (0.1 to 1 mM) for 24 h. In contrast to the 500 nM DOX group, pERK levels were restored in the group co-treated with GSH and suppression of ERK signaling improved hCPCs' survival. Similarly to the previous results, the reduced potency of hCPCs in the 100 nM DOX group, which did not affect cell viability, was ameliorated by co-treatment with GSH (0.1 to 1 mM). Furthermore, GSH was protected against DOX-induced cardiotoxicity in the in vivo model (DOX 20 mg/kg, GSH 100 mg/kg). These results suggest that GSH is a potential therapeutic strategy for DOX-induced cardiotoxicity, which performs its function via ROS reduction and pERK signal regulation.
Collapse
Affiliation(s)
- Eun Ji Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jaewoo Choi
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hye Ji Lim
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sangmi Park
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Vinoth Kumar Rethineswaran
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jong Seong Ha
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jisoo Yun
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Young Joon Hong
- Department of Cardiology, Chonnam National University School of Medicine, Chonnam National University Hospital, Gwangju 61469, Republic of Korea;
| | - Young Jin Choi
- Department of Hemato-Oncology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (E.J.L.); (W.B.J.); (J.C.); (H.J.L.); (S.P.); (V.K.R.); (J.S.H.); (J.Y.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
2
|
Rallapalli S, Guhathakurta S, Korrapati PS. Isolation, growth kinetics, and immunophenotypic characterization of adult human cardiac progenitor cells. J Cell Physiol 2020; 236:1840-1853. [PMID: 33242343 DOI: 10.1002/jcp.29965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 11/10/2022]
Abstract
The discovery of cardiac progenitor cells (CPCs) has raised expectations for the development of cell-based therapy of the heart. Although cell therapy is emerging as a novel treatment for heart failure, several issues still exist concerning an unambiguous definition of the phenotype of CPC types. There is a need to define and validate the methods for the generation of quality CPC populations used in cell therapy applications. Considering the critical roles of cardiac cell progenitors in cellular therapy, we speculate that long term culture might modulate the immunophenotypes of CPCs. Hence, a strategy to validate the isolation and cell culture expansion of cardiac cell populations was devised. Isolation of three subpopulations of human CPCs was done from a single tissue sample using explant, enzymatic isolation, and c-kit+ immunomagnetic sorting methods. The study assessed the effects of ex vivo expansion on proliferation, immunophenotypes, and differentiation of CPCs. Additionally, we report that an explant culture can take over 2 months to achieve similar cell yields, and cell sorting requires a much larger starting population to match this expansion time frame. In comparison, an enzymatic method is expected to yield equivalent quantities of CPCs in 2-3 weeks, notably at a significantly lower cost, which may intensify their use in therapeutic approaches. We determined that ex vivo expansion caused changes in cellular characteristics, and hence propose validated molecular signatures should be established to evaluate the impact of ex vivo expansion for a safe cell therapy product.
Collapse
Affiliation(s)
- Suneel Rallapalli
- Biological Material Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, India
| | | | - Purna S Korrapati
- Biological Material Laboratory, CSIR-Central Leather Research Institute, Adyar, Chennai, India
| |
Collapse
|
3
|
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: 1.2] [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
|
4
|
Therapeutic Cell Protective Role of Histochrome under Oxidative Stress in Human Cardiac Progenitor Cells. Mar Drugs 2019; 17:md17060368. [PMID: 31234277 PMCID: PMC6628112 DOI: 10.3390/md17060368] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022] Open
Abstract
Cardiac progenitor cells (CPCs) are resident stem cells present in a small portion of ischemic hearts and function in repairing the damaged heart tissue. Intense oxidative stress impairs cell metabolism thereby decreasing cell viability. Protecting CPCs from undergoing cellular apoptosis during oxidative stress is crucial in optimizing CPC-based therapy. Histochrome (sodium salt of echinochrome A—a common sea urchin pigment) is an antioxidant drug that has been clinically used as a pharmacologic agent for ischemia/reperfusion injury in Russia. However, the mechanistic effect of histochrome on CPCs has never been reported. We investigated the protective effect of histochrome pretreatment on human CPCs (hCPCs) against hydrogen peroxide (H2O2)-induced oxidative stress. Annexin V/7-aminoactinomycin D (7-AAD) assay revealed that histochrome-treated CPCs showed significant protective effects against H2O2-induced cell death. The anti-apoptotic proteins B-cell lymphoma 2 (Bcl-2) and Bcl-xL were significantly upregulated, whereas the pro-apoptotic proteins BCL2-associated X (Bax), H2O2-induced cleaved caspase-3, and the DNA damage marker, phosphorylated histone (γH2A.X) foci, were significantly downregulated upon histochrome treatment of hCPCs in vitro. Further, prolonged incubation with histochrome alleviated the replicative cellular senescence of hCPCs. In conclusion, we report the protective effect of histochrome against oxidative stress and present the use of a potent and bio-safe cell priming agent as a potential therapeutic strategy in patient-derived hCPCs to treat heart disease.
Collapse
|
5
|
Abstract
A great interest has developed over the last several years in research on interstitial Cajal-like cells (ICLCs), later renamed to telocytes (TCs). Such studies are restricted by diverse limitations. We aimed to critically review (sub)epicardial ICLCs/TCs and to bring forward supplemental immunohistochemical evidence on (sub)epicardial stromal niche inhabitants. We tested the epicardial expressions of CD117/c-kit, CD34, Cytokeratin 7 (CK7), Ki67, Platelet-Derived Growth Factor Receptor (PDGFR)-α and D2-40 in adult human cardiac samples. The mesothelial epicardial cells expressed D2-40, CK7, CD117/c-kit and PDGFR-α. Subepicardial D2-40-positive lymphatic vessels and isolated D2-40-positive and CK7-positive subepicardial cells were also found. Immediate submesothelial spindle-shaped cells expressed Ki-67. Submesothelial stromal cells and endothelial tubes were PDGFR-α-positive and CD34-positive. The expression of CD34 was pan-stromal, so a particular stromal cell type could not be distinguished. The stromal expression of CD117/c-kit was also noted. It seems that epicardial TCs could not be regarded as belonging to a unique cell type until (pre)lymphatic endothelial cells are inadequately excluded. Markers such as CD117/c-kit or CD34 seem to be improper for identifying TCs as a distinctive cell type. Care should be taken when using the immunohistochemical method and histological interpretations, as they may not produce accurate results.
Collapse
|
6
|
Lara-Martínez LA, Gutiérrez-Villegas I, Arenas-Luna VM, Hernández-Gutierrez S. [Stem cells: searching predisposition to cardiac commitment by surface markers expression]. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2018; 88:483-495. [PMID: 29311024 DOI: 10.1016/j.acmx.2017.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 11/30/2017] [Accepted: 12/01/2017] [Indexed: 11/19/2022] Open
Abstract
It is well-known that cardiovascular diseases are the leading cause of death worldwide, and represent an important economic burden to health systems. In an attempt to solve this problem, stem cell therapy has emerged as a therapeutic option. Within the last 20 years, a great variety of stem cells have been used in different myocardial infarction models. Up until now, the use of cardiac stem cells (CSCs) has seemed to be the best option, but the inaccessibility and scarcity of these cells make their use unreliable. Additionally, there is a high risk as they have to be obtained directly from the heart of the patient. Unlike CSCs, adult stem cells originating from bone marrow or adipose tissue, among others, appear to be an attractive option due to their easier accessibility and abundance, but particularly due to the probable existence of cardiac progenitors among their different sub-populations. In this review an analysis is made of the surface markers present in CSCs compared with other adult stem cells. This suggested the pre-existence of cells sharing specific surface markers with CSCs, a predictable immunophenotype present in some cells, although in low proportions, and with a potential of cardiac differentiation that could be similar to CSCs, thus increasing their therapeutic value. This study highlights new perspectives regarding MSCs that would enable some of these sub-populations to be differentiated at cardiac tissue level.
Collapse
Affiliation(s)
- Luis A Lara-Martínez
- Laboratorio de Biología Molecular, Escuela de Medicina, Universidad Panamericana, Ciudad de México, México
| | - Ingrid Gutiérrez-Villegas
- Laboratorio de Biología Molecular, Escuela de Medicina, Universidad Panamericana, Ciudad de México, México
| | - Victor M Arenas-Luna
- Laboratorio de Biología Molecular, Escuela de Medicina, Universidad Panamericana, Ciudad de México, México
| | | |
Collapse
|
7
|
Ledford BT, Simmons J, Chen M, Fan H, Barron C, Liu Z, Van Dyke M, He JQ. Keratose Hydrogels Promote Vascular Smooth Muscle Differentiation from C-kit-Positive Human Cardiac Stem Cells. Stem Cells Dev 2017; 26:888-900. [DOI: 10.1089/scd.2016.0351] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Benjamin T. Ledford
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Jamelle Simmons
- Department of Biomedical Engineering and Mechanics, School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia
| | - Miao Chen
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Huimin Fan
- Research Institute of Heart Failure, Shanghai East Hospital of Tongji University, Shanghai, People's Republic of China
| | - Catherine Barron
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Zhongmin Liu
- Research Institute of Heart Failure, Shanghai East Hospital of Tongji University, Shanghai, People's Republic of China
| | - Mark Van Dyke
- Department of Biomedical Engineering and Mechanics, School of Biomedical Engineering and Sciences, Virginia Tech, Blacksburg, Virginia
| | - Jia-Qiang He
- Department of Biomedical Sciences and Pathobiology, College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| |
Collapse
|
8
|
Ii M. Novel direct reprogramming technique for the generation of culture-expandable cardiac progenitor cells from fibroblasts. Stem Cell Investig 2017; 4:15. [PMID: 28275645 DOI: 10.21037/sci.2017.02.04] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2016] [Accepted: 01/17/2017] [Indexed: 11/06/2022]
Affiliation(s)
- Masaaki Ii
- Division of Research Animal Laboratory and Translational Medicine, Research and Development Center, Osaka Medical College, Osaka, Japan
| |
Collapse
|
9
|
Shi B, Deng W, Long X, Zhao R, Wang Y, Chen W, Xu G, Sheng J, Wang D, Cao S. miR-21 increases c-kit + cardiac stem cell proliferation in vitro through PTEN/PI3K/Akt signaling. PeerJ 2017; 5:e2859. [PMID: 28168101 PMCID: PMC5289448 DOI: 10.7717/peerj.2859] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 12/03/2016] [Indexed: 01/04/2023] Open
Abstract
The low survival rate of cardiac stem cells (CSCs) in the ischemic myocardium is one of the obstacles in ischemic cardiomyopathy cell therapy. The MicroRNA (miR)-21 and one of its target protein, the tensin homolog deleted on chromosome ten (PTEN), contributes to the proliferation of many kinds of tissues and cell types. It is reported that miR-21 promotes proliferation through PTEN/PI3K/Akt pathway, but its effects on c-kit+ CSC remain unclear. The authors hypothesized that miR-21 promotes the proliferation in c-kit + CSC, and evaluated the involvement of PTEN/PI3K/Akt pathway in vitro. miR-21 up-regulation with miR-21 efficiently mimics accelerated cell viability and proliferation in c-kit + CSC, which was evidenced by the CCK-8, EdU and cell cycle analyses. In addition, the over-expression of miR-21 in c-kit + CSCs notably down-regulated the protein expression of PTEN although the mRNA level of PTEN showed little change. Gain-of-function of miR-21 also increased the phosphor-Akt (p-Akt) level. Phen, the selective inhibitor of PTEN, reproduced the pro-proliferation effects of miR-21, while PI3K inhibitor, LY294002, totally attenuated the pro-survival effect of miR-21. These results indicate that miR-21 is efficient in promoting proliferation in c-kit+ CSCs, which is contributed by the PTEN/PI3K/Akt pathway. miR-21 holds the potential to facilitate CSC therapy in ischemic myocardium.
Collapse
Affiliation(s)
- Bei Shi
- Department of Cardiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Wenwen Deng
- Department of Cardiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Xianping Long
- Department of Cardiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Ranzun Zhao
- Department of Cardiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Yan Wang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Wenming Chen
- Department of Cardiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Guanxue Xu
- Department of Cardiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Jin Sheng
- Department of Cardiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Dongmei Wang
- Department of Cardiology, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Song Cao
- Department of Anesthesiology, Zunyi Medical College, Zunyi, Guizhou, China.,Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Zunyi, Guizhou, China
| |
Collapse
|
10
|
Arbatlı S, Aslan GS, Kocabaş F. Stem Cells in Regenerative Cardiology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1079:37-53. [PMID: 29064067 DOI: 10.1007/5584_2017_113] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The common prevalence of heart failure and limitations in its treatment are leading cause of attention and interest towards the induction of cardiac regeneration with novel approaches. Recent studies provide growing evidence regarding bona fide cardiac regeneration post genetic manipulations, administration of stimulatory factors and myocardial injuries in animal models and human studies. To this end, stem cells of different sources have been tested to treat heart failure for the development of cellular therapies. Endogenous and exogenous stem cells sources used in regenerative cardiology have provided a proof of concept and applicability of cellular therapies in myocardial improvement. Recent clinical studies, especially, based on the endogenous cardiac progenitor and stem cells highlighted the possibility to regenerate lost cardiomyocytes in the myocardium. This review discusses emerging concepts in cardiac stem cell therapy, their sources and route of administration, and plausibility of de novo cardiomyocyte formation.
Collapse
Affiliation(s)
- Semih Arbatlı
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
- Department of Biotechnology, Institute of Science, Yeditepe University, Istanbul, Turkey
| | - Galip Servet Aslan
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
- Department of Biotechnology, Institute of Science, Yeditepe University, Istanbul, Turkey
| | - Fatih Kocabaş
- Regenerative Biology Research Laboratory, Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey.
- Department of Biotechnology, Institute of Science, Yeditepe University, Istanbul, Turkey.
| |
Collapse
|
11
|
Liu W, Pu Y, Wang C, Gu C, Zhang X. Expression profiles of long noncoding RNAs in cardiac stem cells under hyperglycemic conditions. Int J Cardiol 2016; 222:933-939. [DOI: 10.1016/j.ijcard.2016.08.028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 08/02/2016] [Indexed: 12/20/2022]
|
12
|
miR-21 Reduces Hydrogen Peroxide-Induced Apoptosis in c-kit + Cardiac Stem Cells In Vitro through PTEN/PI3K/Akt Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:5389181. [PMID: 27803763 PMCID: PMC5075640 DOI: 10.1155/2016/5389181] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 07/06/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022]
Abstract
The low survival rate of cardiac stem cells (CSCs) in the infarcted myocardium hampers cell therapy for ischemic cardiomyopathy. MicroRNA-21 (miR-21) and one of its target proteins, PTEN, contribute to the survival and proliferation of many cell types, but their prosurvival effects in c-kit+ CSC remain unclear. Thus, we hypothesized that miR-21 reduces hydrogen peroxide- (H2O2-) induced apoptosis in c-kit+ CSC and estimated the contribution of PTEN/PI3K/Akt signaling to this oxidative circumstance. miR-21 mimics efficiently reduced H2O2-induced apoptosis in c-kit+ CSC, as evidenced by the downregulation of the proapoptosis proteins caspase-3 and Bax and upregulation of the antiapoptotic Bcl-2. In addition, the gain of function of miR-21 in c-kit+ CSC downregulated the protein level of PTEN although its mRNA level changed slightly; in the meantime, miR-21 overexpression also increased phospho-Akt (p-Akt). The antiapoptotic effects of miR-21 were comparable with Phen (bpV), the selective inhibitor of PTEN, while miR-21 inhibitor or PI3K's inhibitor LY294002 efficiently attenuated the antiapoptotic effect of miR-21. Taken together, these results indicate that the anti-H2O2-induced apoptosis effect of miR-21 in c-kit+ CSC is contributed by PTEN/PI3K/Akt signaling. miR-21 could be a potential molecule to facilitate the c-kit+ CSC therapy in ischemic myocardium.
Collapse
|
13
|
Doxorubicin Regulates Autophagy Signals via Accumulation of Cytosolic Ca 2+ in Human Cardiac Progenitor Cells. Int J Mol Sci 2016; 17:ijms17101680. [PMID: 27735842 PMCID: PMC5085713 DOI: 10.3390/ijms17101680] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/19/2016] [Accepted: 09/28/2016] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (DOXO) is widely used to treat solid tumors. However, its clinical use is limited by side effects including serious cardiotoxicity due to cardiomyocyte damage. Resident cardiac progenitor cells (hCPCs) act as key regulators of homeostasis in myocardial cells. However, little is known about the function of hCPCs in DOXO-induced cardiotoxicity. In this study, we found that DOXO-mediated hCPC toxicity is closely related to calcium-related autophagy signaling and was significantly attenuated by blocking mTOR signaling in human hCPCs. DOXO induced hCPC apoptosis with reduction of SMP30 (regucalcin) and autophagosome marker LC3, as well as remarkable induction of the autophagy-related markers, Beclin-1, APG7, and P62/SQSTM1 and induction of calcium-related molecules, CaM (Calmodulin) and CaMKII (Calmodulin kinase II). The results of an LC3 puncta assay further indicated that DOXO reduced autophagosome formation via accumulation of cytosolic Ca2+. Additionally, DOXO significantly induced mTOR expression in hCPCs, and inhibition of mTOR signaling by rapamycin, a specific inhibitor, rescued DOXO-mediated autophagosome depletion in hCPCs with significant reduction of DOXO-mediated cytosolic Ca2+ accumulation in hCPCs, and restored SMP30 and mTOR expression. Thus, DOXO-mediated hCPC toxicity is linked to Ca2+-related autophagy signaling, and inhibition of mTOR signaling may provide a cardio-protective effect against DOXO-mediated hCPC toxicity.
Collapse
|
14
|
Tian S, Liu Q, Gnatovskiy L, Ma PX, Wang Z. Heart Regeneration with Embryonic Cardiac Progenitor Cells and Cardiac Tissue Engineering. ACTA ACUST UNITED AC 2015; 1. [PMID: 26744736 DOI: 10.19104/jstb.2015.104] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Myocardial infarction (MI) is the leading cause of death worldwide. Recent advances in stem cell research hold great potential for heart tissue regeneration through stem cell-based therapy. While multiple cell types have been transplanted into MI heart in preclinical studies or clinical trials, reduction of scar tissue and restoration of cardiac function have been modest. Several challenges hamper the development and application of stem cell-based therapy for heart regeneration. Application of cardiac progenitor cells (CPCs) and cardiac tissue engineering for cell therapy has shown great promise to repair damaged heart tissue. This review presents an overview of the current applications of embryonic CPCs and the development of cardiac tissue engineering in regeneration of functional cardiac tissue and reduction of side effects for heart regeneration. We aim to highlight the benefits of the cell therapy by application of CPCs and cardiac tissue engineering during heart regeneration.
Collapse
Affiliation(s)
- Shuo Tian
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Qihai Liu
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Leonid Gnatovskiy
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Peter X Ma
- Department of Biologic and Materials Sciences, The University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, The University of Michigan, Ann Arbor, MI 48109, USA; Macromolecular Science and Engineering Center, The University of Michigan, Ann Arbor, MI 48109, USA; Department of Materials Science and Engineering, The University of Michigan, Ann Arbor, MI 48109, USA
| | - Zhong Wang
- Department of Cardiac Surgery, Cardiovascular Center, The University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
15
|
Abstract
This review article discusses the mechanisms of cardiomyogenesis in the adult heart. They include the re-entry of cardiomyocytes into the cell cycle; dedifferentiation of pre-existing cardiomyocytes, which assume an immature replicating cell phenotype; transdifferentiation of hematopoietic stem cells into cardiomyocytes; and cardiomyocytes derived from activation and lineage specification of resident cardiac stem cells. The recognition of the origin of cardiomyocytes is of critical importance for the development of strategies capable of enhancing the growth response of the myocardium; in fact, cell therapy for the decompensated heart has to be based on the acquisition of this fundamental biological knowledge.
Collapse
Affiliation(s)
- Annarosa Leri
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| | - Marcello Rota
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Francesco S Pasqualini
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Polina Goichberg
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Piero Anversa
- From the Departments of Anesthesia and Medicine and Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
16
|
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: 1.2] [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
|
17
|
Guo J, Jie W, Shen Z, Li M, Lan Y, Kong Y, Guo S, Li T, Zheng S. SCF increases cardiac stem cell migration through PI3K/AKT and MMP‑2/‑9 signaling. Int J Mol Med 2014; 34:112-8. [PMID: 24804928 PMCID: PMC4072340 DOI: 10.3892/ijmm.2014.1773] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 04/29/2014] [Indexed: 12/15/2022] Open
Abstract
The transplantation of cardiac stem cells (CSCs) is thought to be responsible for improving the performance of injured heart induced by myocardial infarction (MI). However, the mechanisms involved in the migration of activated CSCs post-MI remain to be clarified. In this study, CSCs were isolated from rat hearts and a cellular migration assay was performed using a 24-well Transwell system. Stem cell factor (SCF) induced CSC migration in a concentration-dependent manner, which could be blocked with an SCF antibody as well as a PI3K/AKT inhibitor, LY294002. Moreover, SCF induced the expression and activity of matrix metalloproteinase (MMP)-2 and MMP-9 in a concentration- and time-dependent manner, as measured by quantitative RT-PCR, western blot analysis and gelatin zymography. Results of western blot analysis revealed phosphorylated AKT was markedly increased in SCF-treated CSCs and that inhibition of SCF/c-Kit signaling or phospho-AKT activity significantly attenuated the SCF-induced expression of MMP-2 and MMP-9. Thus, our results showed that SCF partially mediated CSC migration via the activation of PI3K/AKT/MMP-2/-9 signaling.
Collapse
Affiliation(s)
- Junli Guo
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Wei Jie
- Department of Pathology, School of Basic Medicine Science, Guangdong Medical College, Zhanjiang 524023, P.R. China
| | - Zhihua Shen
- Department of Pathology, School of Basic Medicine Science, Guangdong Medical College, Zhanjiang 524023, P.R. China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China
| | - Youling Lan
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Yueqiong Kong
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Shaoli Guo
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Tianfa Li
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Shaojiang Zheng
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| |
Collapse
|
18
|
Chen Z, Pan X, Yao Y, Yan F, Chen L, Huang R, Ma G. Regulation of c-kit+ progenitor cells by stromal cell derived factor-1α in adult murine heart. Heart Lung Circ 2014; 23:75-81. [PMID: 23891309 DOI: 10.1016/j.hlc.2013.05.652] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/29/2013] [Accepted: 05/30/2013] [Indexed: 01/07/2023]
Abstract
BACKGROUND c-kit-positive cardiac progenitor cells (CPCs) have been proven suitable for stem cell therapy. CPCs marker c-kit and its ligand, the stem cell factor (SCF), are associated with the functions of proliferation and differentiation. In our previous study, we found that stromal cell-derived factor-1α (SDF-1α) could enhance the expression of c-kit. However, the mechanism is unknown. METHODS AND RESULTS CPCs were isolated from adult mouse hearts, and c-kit-positive CPCs were purified by magnetic-activated c-kit cell sorting magnetic beads. The cells were cultured with SDF-1α, c-kit expression was measured by western blotting and qPCR, the proliferation and migration of cells were measured by CCK-8 and transwell assay, DNA methyltransferase (DNMT) mRNA were measured by qPCR, global DNMT activity was measured by DNMT activity assay kit, and DNA methylation was analysed using Sequenom's MassARRAY platform. Results showed that SDF-1α could enhance the expression of c-kit, which results in the promoting of c-kit-positive CPCs proliferation and migration. SDF-1α stimulation inhibited the expression of DNMT1, DNMT3β, and global DNMT activity, which led to significant demethylation in c-kit-positive CPCs. CONCLUSIONS SDF-1α signalling, via CXCR4 activation, up-regulated c-kit expression by inhibiting DNMT1 and DNMT3β expression and global DNMT activity, and by subsequent demethylation of the c-kit gene.
Collapse
Affiliation(s)
- Zhongpu Chen
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Xiaodong Pan
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Fengdi Yan
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Long Chen
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Rong Huang
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu 210009, China.
| |
Collapse
|