1
|
Tan J, Fan L, Li X, Xia LZ, Xu DF, Zhang ZQ, Wang CH, Wu QF, Zhao Y, Li ZM. GnRH antagonist impairs the process of embryo implantation by inhibiting motility of endometrial stromal cells through reducing c-kit expression. Gynecol Endocrinol 2024; 40:2421487. [PMID: 39485323 DOI: 10.1080/09513590.2024.2421487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 09/07/2024] [Accepted: 10/21/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND It has been recognized that the gonadotropin-releasing hormone antagonist (GnRH-ant) protocol has a detrimental effect on clinical outcomes compared to the GnRH agonist (GnRH-a) protocol during in vitro fertilization-fresh embryo transfer (IVF-ET) cycles. However, the related mechanisms were unclear. METHODS A total of 18,561 patients, who underwent fresh IVF-ET cycles in the Center for Assisted Reproduction of Jiangxi Maternal and Child Health Hospital from January 2014 to September 2021, were retrospectively analyzed. The propensity score matching (PSM) technique was used to control for confounding factors between the GnRH-ant and GnRH-a groups. Human endometrial stromal cells (hESCs) were collected for primary culture and treated with relevant receptor antagonists and activators. RT-PCR, Western Blot, immunofluorescence staining, cell migration and adhesion assays, and animal experiments were employed to elucidate the molecular mechanism by which GnRH antagonist affects the migration and adhesion ability of hESCs. RESULTS There was no statistical difference between the two groups in terms of baseline characteristics after matching basal status by propensity score matching. The result showed that the endometrial thickness (10.4 ± 2.35 vs. 11.03 ± 2.61 mm, p < .001) on trigger day was significantly lower in the GnRH-ant group. Compared with the GnRH-a protocol, the implantation rate (39.71% vs. 50.36%, p < .001), biochemical pregnancy rate (64.26% vs. 72.7%, p < .001), clinical pregnancy rate (56.39% vs. 65.24%, p < .001), live birth rate (45.25% vs. 56.1%, p < .001) in the GnRH-ant group were significantly decreased. Contrarily, the rate of early miscarriage in the GnRH-ant group (13.95% vs. 9.04%, p < .001) was higher than in the GnRH-a group. Furthermore, after treating with GnRH-ant, hESCs showed a reduced expression of HOXA10 and MMP-9 proteins, and a weakened migration ability. Subsequently, by establishing the co-culture system of hESCs and JAR trophoblast spheroids, we found that GnRH-ant inhibited the adhesion and invasion ability of trophoblast cells. Moreover, we also found a decreased expression and phosphorylation of c-kit receptor in decidualized hESCs after treating with GnRH-ant. Similar results as observed above were also confirmed when inhibiting the activation of c-kit receptor by imatinib. CONCLUSIONS GnRH-ant could reduce the motility of hESCs by inhibiting the expression and activation of the C-kit receptor, which impaired the process of embryo implantation.
Collapse
Affiliation(s)
- Jun Tan
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Lu Fan
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Xin Li
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Lei-Zhen Xia
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Ding-Fei Xu
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Zhi-Qin Zhang
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Chang-Hua Wang
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Qiong-Fang Wu
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Yan Zhao
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Zeng-Ming Li
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| |
Collapse
|
2
|
Lee W, Lin SL, Chiang CS, Chen JY, Chieng WW, Huang SR, Chang TY, Linju Yen B, Hung MC, Chang KC, Lee HT, Jeng LB, Shyu WC. Role of HIF-1α-Activated IL-22/IL-22R1/Bmi1 Signaling Modulates the Self-Renewal of Cardiac Stem Cells in Acute Myocardial Ischemia. Stem Cell Rev Rep 2024; 20:2194-2214. [PMID: 39264501 PMCID: PMC11554697 DOI: 10.1007/s12015-024-10774-8] [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] [Accepted: 08/06/2024] [Indexed: 09/13/2024]
Abstract
Impaired tissue regeneration negatively impacts on left ventricular (LV) function and remodeling after acute myocardial infarction (AMI). Little is known about the intrinsic regulatory machinery of ischemia-induced endogenous cardiac stem cells (eCSCs) self-renewing divisions after AMI. The interleukin 22 (IL-22)/IL-22 receptor 1 (IL-22R1) pathway has emerged as an important regulator of several cellular processes, including the self-renewal and proliferation of stem cells. However, whether the hypoxic environment could trigger the self-renewal of eCSCs via IL-22/IL-22R1 activation remains unknown. In this study, the upregulation of IL-22R1 occurred due to activation of hypoxia-inducible factor-1α (HIF-1α) under hypoxic and ischemic conditions. Systemic IL-22 administration not only attenuated cardiac remodeling, inflammatory responses, but also promoted eCSC-mediated cardiac repair after AMI. Unbiased RNA microarray analysis showed that the downstream mediator Bmi1 regulated the activation of CSCs. Therefore, the HIF-1α-induced IL-22/IL-22R1/Bmi1 cascade can modulate the proliferation and activation of eCSCs in vitro and in vivo. Collectively, investigating the HIF-1α-activated IL-22/IL-22R1/Bmi1 signaling pathway might offer a new therapeutic strategy for AMI via eCSC-induced cardiac repair.
Collapse
Affiliation(s)
- Wei Lee
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
| | - Syuan-Ling Lin
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Chih-Sheng Chiang
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University (CMU), Taichung, 404, Taiwan
- Neuroscience and Brain Disease Center and New Drug Development Center, CMU, Taichung, 404, Taiwan
| | - Jui-Yu Chen
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Wee-Wei Chieng
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Shu-Rou Huang
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan
| | - Ting-Yu Chang
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
| | - B Linju Yen
- Regenerative Medicine Research Group, Institute of Cellular and System Medicine, National Health Research Institutes (NHRI), Zhunan, 350, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences and Research Centers for Cancer Biology and Molecular Medicine, CMU, Taichung, 404, Taiwan
| | - Kuan-Cheng Chang
- Division of Cardiovascular Medicine, Department of Medicine, CMUH, Taichung, 404, Taiwan
- School of Medicine, CMU, Taichung, 404, Taiwan
| | - Hsu-Tung Lee
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, 404, Taiwan
| | - Long-Bin Jeng
- Cell Therapy Center, China Medical University Hospital (CMUH), Taichung, 404, Taiwan
- Organ Transplantation Center, CMUH, Taichung, 404, Taiwan
| | - Woei-Cherng Shyu
- Translational Medicine Research Center, CMUH, Taichung, 404, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University (CMU), Taichung, 404, Taiwan.
- Neuroscience and Brain Disease Center and New Drug Development Center, CMU, Taichung, 404, Taiwan.
- Department of Neurology, CMUH, Taichung, 404, Taiwan.
- Department of Occupational Therapy, Asia University, No. 2, Yude Rd., North Dist, Taichung City, 404332, Taiwan.
| |
Collapse
|
3
|
Lee WS, Abel ED, Kim J. New Insights into IGF-1 Signaling in the Heart. Physiology (Bethesda) 2024; 39:0. [PMID: 38713091 DOI: 10.1152/physiol.00003.2024] [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: 01/10/2024] [Revised: 04/24/2024] [Accepted: 05/04/2024] [Indexed: 05/08/2024] Open
Abstract
Insulin-like growth factor-1 (IGF-1) signaling has multiple physiological roles in cellular growth, metabolism, and aging. Myocardial hypertrophy, cell death, senescence, fibrosis, and electrical remodeling are hallmarks of various heart diseases and contribute to the progression of heart failure. This review highlights the critical role of IGF-1 and its cognate receptor in cardiac hypertrophy, aging, and remodeling.
Collapse
Affiliation(s)
- Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - E Dale Abel
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
4
|
Dergilev K, Tsokolaeva Z, Goltseva Y, Beloglazova I, Ratner E, Parfyonova Y. Urokinase-Type Plasminogen Activator Receptor Regulates Prosurvival and Angiogenic Properties of Cardiac Mesenchymal Stromal Cells. Int J Mol Sci 2023; 24:15554. [PMID: 37958542 PMCID: PMC10650341 DOI: 10.3390/ijms242115554] [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: 07/24/2023] [Revised: 09/29/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
One of the largest challenges to the implementation of cardiac cell therapy is identifying selective reparative targets to enhance stem/progenitor cell therapeutic efficacy. In this work, we hypothesized that such a target could be an urokinase-type plasminogen activator receptor (uPAR)-a glycosyl-phosphatidyl-inositol-anchored membrane protein, interacting with urokinase. uPAR is able to form complexes with various transmembrane proteins such as integrins, activating intracellular signaling pathway and thus regulating multiple cell functions. We focused on studying the CD117+ population of cardiac mesenchymal progenitor cells (MPCs), expressing uPAR on their surface. It was found that the number of CD117+ MPCs in the heart of the uPAR-/- mice is lower, as well as their ability to proliferate in vitro compared with cells from wild-type animals. Knockdown of uPAR in CD117+ MPCs of wild-type animals was accompanied by a decrease in survival rate and Akt signaling pathway activity and by an increase in the level of caspase activity in these cells. That suggests the role of uPAR in supporting cell survival. After intramyocardial transplantation of uPAR(-) MPCs, reduced cell retention and angiogenesis stimulation were observed in mice with myocardial infarction model compared to uPAR(+) cells transplantation. Taken together, the present results appear to prove a novel mechanism of uPAR action in maintaining the survival and angiogenic properties of CD117+ MPCs. These results emphasize the importance of the uPAR as a potential pharmacological target for the regulation of reparative properties of myocardial mesenchymal progenitor cells.
Collapse
Affiliation(s)
- Konstantin Dergilev
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
| | - Zoya Tsokolaeva
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Yulia Goltseva
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
| | - Irina Beloglazova
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
| | - Elizaveta Ratner
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
| | - Yelena Parfyonova
- Institute of Experimental Cardiology Named after Academician V.N. Smirnov, Federal State Budgetary Institution National Medical Research Center of Cardiology Named after Academician E.I. Chazov, Ministry of Health of the Russian Federation, 121552 Moscow, Russia; (K.D.)
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| |
Collapse
|
5
|
Khedkar HN, Chen LC, Kuo YC, Wu ATH, Huang HS. Multi-Omics Identification of Genetic Alterations in Head and Neck Squamous Cell Carcinoma and Therapeutic Efficacy of HNC018 as a Novel Multi-Target Agent for c-MET/STAT3/AKT Signaling Axis. Int J Mol Sci 2023; 24:10247. [PMID: 37373393 DOI: 10.3390/ijms241210247] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/07/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
Amongst the most prevalent malignancies worldwide, head and neck squamous cell carcinoma (HNSCC) is characterized by high morbidity and mortality. The failure of standard treatment modalities, such as surgery, radiotherapy, and chemotherapy, demands the need for in-depth understanding of the complex signaling networks involved in the development of treatment resistance. A tumor's invasive growth and high levels of intrinsic or acquired treatment resistance are the primary causes of treatment failure. This may be a result of the presence of HNSCC's cancer stem cells, which are known to have self-renewing capabilities that result in therapeutic resistance. Using bioinformatics methods, we discovered that elevated expressions of MET, STAT3, and AKT were associated with poor overall survival in HNSCC patients. We then evaluated the therapeutic potential of our newly synthesized small molecule HNC018 towards its potential as a novel anticancer drug. Our computer-aided structure characterization and target identification study predicted that HNC018 could target these oncogenic markers implicated in HNSCC. Subsequently, the HNC018 has demonstrated its anti-proliferative and anticancer activities towards the head and neck squamous cell carcinoma cell lines, along with displaying the stronger binding affinities towards the MET, STAT3, and AKT than the standard drug cisplatin. Reduction in the clonogenic and tumor-sphere-forming ability displays HNC018's role in decreasing the tumorigenicity. Importantly, an vivo study has shown a significant delay in tumor growth in HNC018 alone or in combination with cisplatin-treated xenograft mice model. Collectively with our findings, HNC018 highlights the desirable properties of a drug-like candidate and could be considered as a novel small molecule for treating head and neck squamous cell carcinoma.
Collapse
Affiliation(s)
- Harshita Nivrutti Khedkar
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, and Academia Sinica, Taipei 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| | - Lung-Ching Chen
- Division of Cardiology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 11101, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei 24205, Taiwan
| | - Yu-Cheng Kuo
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
| | - Alexander T H Wu
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Taipei Heart Institute (THI), Taipei Medical University, Taipei 11031, Taiwan
- Clinical Research Center, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Centre, Taipei 11490, Taiwan
| | - Hsu-Shan Huang
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, and Academia Sinica, Taipei 11031, Taiwan
- Graduate Institute for Cancer Biology & Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Graduate Institute of Medical Sciences, National Defense Medical Centre, Taipei 11490, Taiwan
- School of Pharmacy, National Defense Medical Centre, Taipei 11490, Taiwan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
6
|
Kanda M, Nagai T, Kondo N, Matsuura K, Akazawa H, Komuro I, Kobayashi Y. Pericardial Grafting of Cardiac Progenitor Cells in Self-Assembling Peptide Scaffold Improves Cardiac Function After Myocardial Infarction. Cell Transplant 2023; 32:9636897231174078. [PMID: 37191272 PMCID: PMC10192947 DOI: 10.1177/09636897231174078] [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: 03/17/2022] [Revised: 04/03/2023] [Accepted: 04/19/2023] [Indexed: 05/17/2023] Open
Abstract
Many studies have explored cardiac progenitor cell (CPC) therapy for heart disease. However, optimal scaffolds are needed to ensure the engraftment of transplanted cells. We produced a three-dimensional hydrogel scaffold (CPC-PRGmx) in which high-viability CPCs were cultured for up to 8 weeks. CPC-PRGmx contained an RGD peptide-conjugated self-assembling peptide with insulin-like growth factor-1 (IGF-1). Immediately after creating myocardial infarction (MI), we transplanted CPC-PRGmx into the pericardial space on to the surface of the MI area. Four weeks after transplantation, red fluorescent protein-expressing CPCs and in situ hybridization analysis in sex-mismatched transplantations revealed the engraftment of CPCs in the transplanted scaffold (which was cellularized with host cells). The average scar area of the CPC-PRGmx-treated group was significantly smaller than that of the non-treated group (CPC-PRGmx-treated group = 46 ± 5.1%, non-treated MI group = 59 ± 4.5%; p < 0.05). Echocardiography showed that the transplantation of CPC-PRGmx improved cardiac function and attenuated cardiac remodeling after MI. The transplantation of CPCs-PRGmx promoted angiogenesis and inhibited apoptosis, compared to the untreated MI group. CPCs-PRGmx secreted more vascular endothelial growth factor than CPCs cultured on two-dimensional dishes. Genetic fate mapping revealed that CPC-PRGmx-treated mice had more regenerated cardiomyocytes than non-treated mice in the MI area (CPC-PRGmx-treated group = 0.98 ± 0.25%, non-treated MI group = 0.25 ± 0.04%; p < 0.05). Our findings reveal the therapeutic potential of epicardial-transplanted CPC-PRGmx. Its beneficial effects may be mediated by sustainable cell viability, paracrine function, and the enhancement of de novo cardiomyogenesis.
Collapse
Affiliation(s)
- Masato Kanda
- Department of Cardiovascular Medicine,
Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshio Nagai
- Department of Cardiology, Chemotherapy
Research Institute, KAKEN Hospital, International University of Health and Welfare,
Ichikawa-shi, Japan
| | - Naomichi Kondo
- Department of Cardiovascular Medicine,
Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Katsuhisa Matsuura
- Institute of Advanced Biomedical
Engineering and Science, Tokyo Women’s Medical University, Tokyo, Japan
- Department of Cardiology, Tokyo Women’s
Medical University, Tokyo, Japan
| | - Hiroshi Akazawa
- Department of Cardiovascular Medicine,
Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine,
Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yoshio Kobayashi
- Department of Cardiovascular Medicine,
Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
7
|
Cagnin S, Brugnaro M, Millino C, Pacchioni B, Troiano C, Di Sante M, Kaludercic N. Monoamine Oxidase-Dependent Pro-Survival Signaling in Diabetic Hearts Is Mediated by miRNAs. Cells 2022; 11:2697. [PMID: 36078109 PMCID: PMC9454570 DOI: 10.3390/cells11172697] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 10/05/2023] Open
Abstract
Diabetes leads to cardiomyopathy and heart failure, the leading cause of death for diabetic patients. Monoamine oxidase (MAO) inhibition in diabetic cardiomyopathy prevents oxidative stress, mitochondrial and endoplasmic reticulum stress and the development of diastolic dysfunction. However, it is unclear whether, in addition to the direct effects exerted on the mitochondria, MAO activity is able to post-transcriptionally regulate cardiomyocyte function and survival in diabetes. To this aim, we performed gene and miRNA expression profiling in cardiac tissue from streptozotocin-treated mice (model of type 1 diabetes (T1D)), administered with either vehicle or MAOs inhibitor pargyline for 12 weeks. We found that inhibition of MAO activity in T1D hearts leads to profound transcriptomic changes, affecting autophagy and pro-survival pathways activation. MAO activity in T1D hearts increased miR-133a-3p, -193a-3p and -27a-3p expression. These miRNAs target insulin-like growth factor receptor 1 (Igf1r), growth factor receptor bound protein 10 and inositol polyphosphate 4 phosphatase type 1A, respectively, all components of the IGF1R/PI3K/AKT signaling pathway. Indeed, AKT activation was significantly downregulated in T1D hearts, whereas MAO inhibition restored the activation of this pro-survival pathway. The present study provides an important link between MAO activity, transcriptomic changes and activation of pro-survival signaling and autophagy in diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Stefano Cagnin
- Department of Biology, University of Padova, 35131 Padova, Italy
- CIR-Myo Myology Center, University of Padova, 35131 Padova, Italy
| | - Marco Brugnaro
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Caterina Millino
- Department of Biology, University of Padova, 35131 Padova, Italy
| | | | - Carmen Troiano
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Moises Di Sante
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Nina Kaludercic
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Neuroscience Institute, National Research Council of Italy (CNR), 35131 Padova, Italy
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127 Padova, Italy
| |
Collapse
|
8
|
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
|
9
|
Sharma A, Gupta S, Archana S, Verma RS. Emerging Trends in Mesenchymal Stem Cells Applications for Cardiac Regenerative Therapy: Current Status and Advances. Stem Cell Rev Rep 2022; 18:1546-1602. [PMID: 35122226 DOI: 10.1007/s12015-021-10314-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2021] [Indexed: 12/29/2022]
Abstract
Irreversible myocardium infarction is one of the leading causes of cardiovascular disease (CVD) related death and its quantum is expected to grow in coming years. Pharmacological intervention has been at the forefront to ameliorate injury-related morbidity and mortality. However, its outcomes are highly skewed. As an alternative, stem cell-based tissue engineering/regenerative medicine has been explored quite extensively to regenerate the damaged myocardium. The therapeutic modality that has been most widely studied both preclinically and clinically is based on adult multipotent mesenchymal stem cells (MSC) delivered to the injured heart. However, there is debate over the mechanistic therapeutic role of MSC in generating functional beating cardiomyocytes. This review intends to emphasize the role and use of MSC in cardiac regenerative therapy (CRT). We have elucidated in detail, the various aspects related to the history and progress of MSC use in cardiac tissue engineering and its multiple strategies to drive cardiomyogenesis. We have further discussed with a focus on the various therapeutic mechanism uncovered in recent times that has a significant role in ameliorating heart-related problems. We reviewed recent and advanced technologies using MSC to develop/create tissue construct for use in cardiac regenerative therapy. Finally, we have provided the latest update on the usage of MSC in clinical trials and discussed the outcome of such studies in realizing the full potential of MSC use in clinical management of cardiac injury as a cellular therapy module.
Collapse
Affiliation(s)
- Akriti Sharma
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, 600036, Tamil Nadu, India
| | - Santosh Gupta
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, 600036, Tamil Nadu, India
| | - S Archana
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, 600036, Tamil Nadu, India
| | - Rama Shanker Verma
- Stem Cell and Molecular Biology Laboratory, Bhupat and Jyoti Mehta School of Biosciences, Department of Biotechnology, Indian Institute of Technology-Madras, Chennai, 600036, Tamil Nadu, India.
| |
Collapse
|
10
|
Desole C, Gallo S, Vitacolonna A, Vigna E, Basilico C, Montarolo F, Zuppini F, Casanova E, Miggiano R, Ferraris DM, Bertolotto A, Comoglio PM, Crepaldi T. Engineering, Characterization, and Biological Evaluation of an Antibody Targeting the HGF Receptor. Front Immunol 2021; 12:775151. [PMID: 34925346 PMCID: PMC8679783 DOI: 10.3389/fimmu.2021.775151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The Hepatocyte growth factor (HGF) and its receptor (MET) promote several physiological activities such as tissue regeneration and protection from cell injury of epithelial, endothelial, neuronal and muscle cells. The therapeutic potential of MET activation has been scrutinized in the treatment of acute tissue injury, chronic inflammation, such as renal fibrosis and multiple sclerosis (MS), cardiovascular and neurodegenerative diseases. On the other hand, the HGF-MET signaling pathway may be caught by cancer cells and turned to work for invasion, metastasis, and drug resistance in the tumor microenvironment. Here, we engineered a recombinant antibody (RDO24) and two derived fragments, binding the extracellular domain (ECD) of the MET protein. The antibody binds with high affinity (8 nM) to MET ECD and does not cross-react with the closely related receptors RON nor with Semaphorin 4D. Deletion mapping studies and computational modeling show that RDO24 binds to the structure bent on the Plexin-Semaphorin-Integrin (PSI) domain, implicating the PSI domain in its binding to MET. The intact RDO24 antibody and the bivalent Fab2, but not the monovalent Fab induce MET auto-phosphorylation, mimicking the mechanism of action of HGF that activates the receptor by dimerization. Accordingly, the bivalent recombinant molecules induce HGF biological responses, such as cell migration and wound healing, behaving as MET agonists of therapeutic interest in regenerative medicine. In vivo administration of RDO24 in the murine model of MS, represented by experimental autoimmune encephalomyelitis (EAE), delays the EAE onset, mitigates the early clinical symptoms, and reduces inflammatory infiltrates. Altogether, these results suggest that engineered RDO24 antibody may be beneficial in multiple sclerosis and possibly other types of inflammatory disorders.
Collapse
Affiliation(s)
- Claudia Desole
- Department of Oncology, University of Turin, Candiolo, Italy
| | - Simona Gallo
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Annapia Vitacolonna
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Elisa Vigna
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | | | - Francesca Montarolo
- Neuroscience Institute Cavalieri Ottolenghi (NICO), Orbassano, Italy.,Department of Molecular Biotechnology and Health Sciences, University of Turin, Torino, Italy
| | | | | | - Riccardo Miggiano
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy.,IXTAL srl, Novara, Italy
| | - Davide Maria Ferraris
- Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy.,IXTAL srl, Novara, Italy
| | | | | | - Tiziana Crepaldi
- Department of Oncology, University of Turin, Candiolo, Italy.,Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| |
Collapse
|
11
|
Yusuf AM, Qaisar R, Al-Tamimi AO, Jayakumar MN, Woodgett JR, Koch WJ, Ahmad F. Cardiomyocyte-GSK-3β deficiency induces cardiac progenitor cell proliferation in the ischemic heart through paracrine mechanisms. J Cell Physiol 2021; 237:1804-1817. [PMID: 34812500 DOI: 10.1002/jcp.30644] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 10/28/2021] [Accepted: 11/09/2021] [Indexed: 12/15/2022]
Abstract
Cardiomyopathy is an irreparable loss and novel strategies are needed to induce resident cardiac progenitor cell (CPC) proliferation in situ to enhance the possibility of cardiac regeneration. Here, we sought to identify the potential roles of glycogen synthase kinase-3β (GSK-3β), a critical regulator of cell proliferation and differentiation, in CPC proliferation post-myocardial infarction (MI). Cardiomyocyte-specific conditional GSK-3β knockout (cKO) and littermate control mice were employed and challenged with MI. Though cardiac left ventricular chamber dimension and contractile functions were comparable at 2 weeks post-MI, cKO mice displayed significantly preserved LV chamber and contractile function versus control mice at 4 weeks post-MI. Consistent with protective phenotypes, an increased percentage of c-kit-positive cells (KPCs) were observed in the cKO hearts at 4 and 6 weeks post-MI which was accompanied by increased levels of cardiomyocyte proliferation. Further analysis revealed that the observed increased number of KPCs in the ischemic cKO hearts was mainly from a cardiac lineage, as the majority of identified KPCs were negative for the hematopoietic lineage marker, CD45. Mechanistically, cardiomyocyte-GSK-3β profoundly suppresses the expression and secretion of growth factors, including basic-fibroblast growth factor, angiopoietin-2, erythropoietin, stem cell factor, platelet-derived growth factor-BB, granulocyte colony-stimulating factor, and vascular endothelial growth factor, post-hypoxia. In conclusion, our findings strongly suggest that loss of cardiomyocyte-GSK-3β promotes cardiomyocyte and resident CPC proliferation post-MI. The induction of cardiomyocyte and CPC proliferation in the ischemic cKO hearts is potentially regulated by autocrine and paracrine signaling governed by dysregulated growth factors post-MI. A strategy to inhibit cardiomyocyte-GSK-3β could be helpful for the promotion of in situ cardiac regeneration post-ischemic injury.
Collapse
Affiliation(s)
- Ayesha M Yusuf
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE.,Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - Rizwan Qaisar
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE.,Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - Abaher O Al-Tamimi
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE.,Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - Manju Nidagodu Jayakumar
- Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE
| | - James R Woodgett
- Department of Medical Biophysics, Sinai Health System, Lunenfeld-Tanenbaum Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Walter J Koch
- Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Firdos Ahmad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, UAE.,Cardiovascular Research Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah, UAE.,Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
12
|
Park Y, Ji ST, Yong U, Das S, Jang WB, Ahn G, Kwon SM, Jang J. 3D bioprinted tissue-specific spheroidal multicellular microarchitectures for advanced cell therapy. Biofabrication 2021; 13. [PMID: 34433153 DOI: 10.1088/1758-5090/ac212e] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/25/2021] [Indexed: 01/05/2023]
Abstract
Intercellular interaction is the most crucial factor in promoting cell viability and functionality in an engineered tissue system. Of the various shapes available for cell-laden constructs, spheroidal multicellular microarchitectures (SMMs) have been introduced as building blocks and injectable cell carriers with substantial cell-cell and cell-extracellular matrix (ECM) interactions. Here, we developed a precise and expeditious SMM printing method that can create a tissue-specific microenvironment and thus be potentially useful for cell therapy. This printing strategy is designed to manufacture SMMs fabricated with optimal bioink blended with decellularized ECM and alginate to enhance the functional performance of the encapsulated cells. Experimental results showed that the proposed method allowed for size controllability and mass production of SMMs with high cell viability. Moreover, SMMs co-cultured with endothelial cells promoted lineage-specific maturation and increased functionality compared to monocultured SMMs. Overall, it was concluded that SMMs have the potential for use in cell therapy due to their high cell retention and proliferation rate compared to single-cell injection, particularly for efficient tissue regeneration after myocardial infarction. This study suggests that utilizing microextrusion-based 3D bioprinting technology to encapsulate cells in cell-niche-standardized SMMs can expand the range of possible applications.
Collapse
Affiliation(s)
- Yejin Park
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 37673, Republic of Korea
| | - Seung Taek Ji
- Stem Cell Research Center, Medical Research Institute, Pusan National University School of Medicine, Yangsan, Kyungnam 50612, Republic of Korea
| | - Uijung Yong
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 37673, Republic of Korea
| | - Sanskrita Das
- Department of Biomedical Engineering, Emory University, Atlanta, GA 30322, United States of America
| | - Woong Bi Jang
- Stem Cell Research Center, Medical Research Institute, Pusan National University School of Medicine, Yangsan, Kyungnam 50612, Republic of Korea
| | - Geunseon Ahn
- Research Institute, Sphebio Co., Ltd, Pohang, Kyungbuk 37666, Republic of Korea
| | - Sang-Mo Kwon
- Stem Cell Research Center, Medical Research Institute, Pusan National University School of Medicine, Yangsan, Kyungnam 50612, Republic of Korea
| | - Jinah Jang
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 37673, Republic of Korea.,School of Interdisciplinary Bioscience and Bioengineering, POSTECH, Pohang, Kyungbuk 37673, Republic of Korea.,Department of Mechanical Engineering, POSTECH, Pohang, Kyungbuk 37673, Republic of Korea
| |
Collapse
|
13
|
Ivankovic I, Déan-Ben XL, Haas H, Kimm MA, Wildgruber M, Razansky D. Volumetric Optoacoustic Tomography Differentiates Myocardial Remodelling. Mol Imaging Biol 2021; 22:1235-1243. [PMID: 32394284 DOI: 10.1007/s11307-020-01498-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Myocardial healing following myocardial infarction (MI) is a complex process that is yet to be fully understood. Clinical attempts in regeneration of the injured myocardium using cardiac stem cells faced major challenges, calling for a better understanding of the processes involved at a more basic level in order to foster translation. PROCEDURES We examined the feasibility of volumetric optoacoustic tomography (VOT) in studying healing of the myocardium in different models of MI, including permanent occlusion (PO) of the left coronary artery, temporary occlusion (ischemia-reperfusion-I/R) and infarcted c-kit mutants, a genetic mouse model with impaired cardiac healing. Murine hearts were imaged at 100 Hz frame rate using 800 nm excitation wavelength, corresponding to the peak absorption of indocyanine green (ICG) in plasma and the isosbestic point of haemoglobin. RESULTS The non-invasive real-time volumetric imaging capabilities of VOT have allowed the detection of significant variations in the pulmonary transit time (PTT), a parameter affected by MI, across different murine models. Upon intravenous injection of ICG, we were able to track alterations in cardiac perfusion in I/R models, which were absent in wild-type (wt) PO or kitW/kitW-v PO mice. The wt-PO and I/R models further exhibited irregularities in their cardiac cycles. CONCLUSIONS Clear differences in the PTT, ICG perfusion and cardiac cycle patterns were identified between the different models and days post MI. Overall, the results highlight the unique capacity of VOT for multi-parametric characterization of morphological and functional changes in murine models of MI.
Collapse
Affiliation(s)
- Ivana Ivankovic
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Xosé Luís Déan-Ben
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Helena Haas
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar Technical University of Munich, Munich, Germany
| | - Melanie A Kimm
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar Technical University of Munich, Munich, Germany
| | - Moritz Wildgruber
- Department of Diagnostic and Interventional Radiology, Klinikum Rechts der Isar Technical University of Munich, Munich, Germany
- Translational Research Imaging Center, Department of Clinical Radiology, Universitätsklinikum Münster, Munster, Germany
| | - Daniel Razansky
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
14
|
Bektik E, Fu JD. Production of Cardiomyocyte-Like Cells by Fibroblast Reprogramming with Defined Factors. Methods Mol Biol 2021; 2239:33-46. [PMID: 33226611 DOI: 10.1007/978-1-0716-1084-8_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Over the last decade, great achievements have been made in the field of direct epigenetic reprogramming, which converts one type of adult somatic cells into another type of differentiated cells, such as direct reprogramming of fibroblasts into cardiomyocytes, without passage through an undifferentiated pluripotent stage. Discovery of direct cardiac reprogramming offers a promising therapeutic strategy to prevent/attenuate cardiac fibrotic remodeling in a diseased heart. Furthermore, in vitro reprogramming of fibroblasts into cardiomyocyte-like cells provides new avenues to conduct basic mechanistic studies, to test drugs, and to model cardiac diseases in a dish. Here, we describe a detailed step-by-step protocol for in vitro production of induced cardiomyocyte-like cells (iCMs) from fibroblasts. The related procedures include high-quality fibroblast isolation of different origins (neonatal cardiac, tail-tip, and adult cardiac fibroblasts), retroviral preparation of reprogramming factors, and iCM generation by fibroblast reprogramming via retroviral transduction of Gata4, Mef2c, and Tbx5. A detailed written protocol will help many other laboratories, inexperienced in this area, to use and further improve this technology in their studies of cardiac regenerative medicine.
Collapse
Affiliation(s)
- Emre Bektik
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA
| | - Ji-Dong Fu
- The Dorothy M. Davis Heart and Lung Research Institute, Frick Center for Heart Failure and Arrhythmia, The Ohio State University Wexner Medical Center, Columbus, OH, USA. .,Department of Physiology and Cell Biology, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
15
|
Ayad O, Al Sayed ZR, Sebille S, Magaud C, Chapotte-Baldacci CA, Jayle C, Faivre JF, Gaborit N, Chatelier A, Bois P. In vitro differentiation of W8B2 + human cardiac stem cells: gene expression of ionic channels and spontaneous calcium activity. Cell Mol Biol Lett 2020; 25:50. [PMID: 33292162 PMCID: PMC7646077 DOI: 10.1186/s11658-020-00242-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/29/2020] [Indexed: 11/18/2022] Open
Abstract
Background Human cardiac stem cells expressing the W8B2 marker (W8B2+ CSCs) were recently identified and proposed as a new model of multipotent CSCs capable of differentiating into smooth muscle cells, endothelial cells and immature myocytes. Nevertheless, no characterization of ion channel or calcium activity during the differentiation of these stem cells has been reported. Methods The objectives of this study were thus to analyze (using the TaqMan Low-Density Array technique) the gene profile of W8B2+ CSCs pertaining to the regulation of ion channels, transporters and other players involved in the calcium homeostasis of these cells. We also analyzed spontaneous calcium activity (via the GCaMP calcium probe) during the in vitro differentiation of W8B2+ CSCs into cardiac myocytes. Results Our results show an entirely different electrophysiological genomic profile between W8B2+ CSCs before and after differentiation. Some specific nodal genes, such as Tbx3, HCN, ICaT, L, KV, and NCX, are overexpressed after this differentiation. In addition, we reveal spontaneous calcium activity or a calcium clock whose kinetics change during the differentiation process. A pharmacological study carried out on differentiated W8B2+ CSCs showed that the NCX exchanger and IP3 stores play a fundamental role in the generation of these calcium oscillations. Conclusions Taken together, the present results provide important information on ion channel expression and intrinsic calcium dynamics during the differentiation process of stem cells expressing the W8B2 marker.
Collapse
Affiliation(s)
- Oualid Ayad
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Zeina R Al Sayed
- CNRS, INSERM, l'institut du thorax, Université de Nantes, 44000, Nantes, France
| | - Stéphane Sebille
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Christophe Magaud
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | | | - Christophe Jayle
- CHU of Poitiers chirurgie cardiaque et thoracique, , Poitiers Cedex 09, France
| | - Jean-François Faivre
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Nathalie Gaborit
- CNRS, INSERM, l'institut du thorax, Université de Nantes, 44000, Nantes, France
| | - Aurélien Chatelier
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France
| | - Patrick Bois
- University of Poitiers Signalisation et Transports Ioniques Membranaires, EA7349, Poitiers Cedex 09, France.
| |
Collapse
|
16
|
Bianchi VE. Caloric restriction in heart failure: A systematic review. Clin Nutr ESPEN 2020; 38:50-60. [PMID: 32690177 DOI: 10.1016/j.clnesp.2020.04.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/17/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Nutrition exerts a determinant role in maintaining cardiac function, regulating insulin and mitochondrial efficiency, that are essential to support energy production for contractility. In patients with heart failure (HF), myocardial tissue efficiency is reduced because of decreased mitochondrial oxidative capacity. In HF conditions, cardiomyocytes shift toward glucose and a reduction in fatty acid utilization. Calorie restriction induces weight loss in obese patients and can be beneficial in some HF patients, although this has generated some controversy. This study aims to evaluate the impact of the CR diet on myocardial efficiency in HF patients. METHODS On Pubmed and Embase, articles related to the keywords: "chronic heart failure" with "diet," "nutrition," "insulin resistance," and "caloric restriction" have been searched, Studies, including exercise or food supplementation, were excluded. RESULTS The retrieved articles showed that weight loss, through the activation of insulin and various kinase pathways, regulates the efficiency of myocardial tissue. In contrast, insulin resistance represents a strong cardiovascular risk factor that reduces myocardial function. CONCLUSION CR diet represents the first therapy in overweight HF patients, both with preserved ejection fraction (HFpEF) and with reduced ejection fraction (HFrHF) because reducing body fat, the myocardial function increased. Insulin activity is the critical hormone that regulates mitochondrial function and cardiac efficiency. However, a severely restricted diet may represent a severe risk factor correlated with all-cause mortality, particularly in underweight HF patients. Long-term studies conducted on large populations are necessary to evaluate the effects of CR on myocardial function in HF patients.
Collapse
|
17
|
The Long-Lasting Protective Effect of HGF in Cardiomyoblasts Exposed to Doxorubicin Requires a Positive Feed-Forward Loop Mediated by Erk1,2-Timp1-Stat3. Int J Mol Sci 2020; 21:ijms21155258. [PMID: 32722178 PMCID: PMC7432797 DOI: 10.3390/ijms21155258] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 12/16/2022] Open
Abstract
Previous studies showed that the hepatocyte growth factor (HGF)–Met receptor axis plays long-lasting cardioprotection against doxorubicin anti-cancer therapy. Here, we explored the mechanism(s) underlying the HGF protective effect. DNA damage was monitored by histone H2AX phosphorylation and apoptosis by proteolytic cleavage of caspase 3. In doxorubicin-treated H9c2 cardiomyoblasts, the long-lasting cardioprotection is mediated by activation of the Ras/Raf/Mek/Erk (extracellular signal-regulated kinase 1,2) signaling pathway and requires Stat3 (signal transducer and activator of transcription 3) activation. The HGF protection was abrogated by the Erk1,2 inhibitor, PD98059. This translated into reduced Y705 phosphorylation and impaired nuclear translocation of Stat3, showing crosstalk between Erk1,2 and Stat3 signaling. An array of 29 cytokines, known to activate Stat3, was interrogated to identify the molecule(s) linking the two pathways. The analysis showed a selective increase in expression of the tissue inhibitor of metalloproteinases-1 (Timp1). Consistently, inhibition in cardiomyoblasts of Timp1 translation by siRNAs blunted both Stat3 activation and the cardioprotective effect of HGF. Thus, Timp1 is responsible for the generation of a feed-forward loop of Stat3 activation and helps cardiomyocytes to survive during the genotoxic stress induced by anthracyclines.
Collapse
|
18
|
Ceausu Z, Socea B, Dimitriu MCT, Predescu D, Constantin VD, Bacalbaşa N, Cîrstoveanu C, Costache M, Ceausu M. Dormant cardiac stem cells: A promising tool in cardiac regeneration. Exp Ther Med 2020; 20:3452-3457. [PMID: 32905130 PMCID: PMC7465489 DOI: 10.3892/etm.2020.9015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Stem cells represent an old niche with various new potential therapeutics. Besides drug treatment, reperfusion procedures and surgical revascularization, stem cell therapy could be a good option in ischemic cardiac diseases. A study was performed on a small group of cases who died of cardiac arrhythmia secondary to scarring myocardial infarctions. Tissue cardiac samples were taken from these cases (from the anterior and lateral wall of the left ventricle), for microscopy examination, in order to investigate the presence of cardiac stem cells (CSC). Multiple series of histological sections were also performed and examined, along with immunohistochemical analysis (IHC). The cells were identified in close contact with the residual ischemic cardiomyocytes, in the proximity of the myocardial collagenous scar, in old myocardial infarctions. They were activated by hypoxic ischemia and were influenced by the capillary microvascular density and the interstitial micro-environment conditions. In chronic intermittent ischemia they seem to turn themselves from dormant quiescent cells into activated progenitor committed cells.
Collapse
Affiliation(s)
- Zenaida Ceausu
- Pathology Department, 'Sf. Pantelimon' Emergency Clinical Hospital, 021659 Bucharest, Romania
| | - Bogdan Socea
- Department of Surgery, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Surgery, 'Sf. Pantelimon' Emergency Clinical Hospital, 021659 Bucharest, Romania
| | - Mihai C T Dimitriu
- Department of Obstetrics and Gynecology, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Obstetrics and Gynecology, 'Sf. Pantelimon' Emergency Clinical Hospital, 021659 Bucharest, Romania
| | - Dragoş Predescu
- Department of Surgery, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Surgery, 'Sf. Maria' Hospital, 011172 Bucharest, Romania
| | - Vlad D Constantin
- Department of Surgery, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Surgery, 'Sf. Pantelimon' Emergency Clinical Hospital, 021659 Bucharest, Romania
| | - Nicolae Bacalbaşa
- Department of Obstetrics and Gynecology, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Obstetrics and Gynecology, 'Dr. I. Cantacuzino' Clinical Hospital, 020475 Bucharest, Romania
| | - Cătălin Cîrstoveanu
- Pediatrics Department, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Pediatrics Department, 'Maria Sklodowska Curie' Emergency Clinical Hospital for Children, 050831 Bucharest, Romania
| | - Mariana Costache
- Pathology Department, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Pathology Department, University Emergency Hospital, 050098 Bucharest, Romania
| | - Mihai Ceausu
- Pathology Department, 'Carol Davila' University of Medicine and Pharmacy, 020021 Bucharest, Romania.,Department of Pathology, 'Mina Minovici' National Institute of Legal Medicine, 042122 Bucharest, Romania
| |
Collapse
|
19
|
Gallo S, Spilinga M, Albano R, Ferrauto G, Di Gregorio E, Casanova E, Balmativola D, Bonzano A, Boccaccio C, Sapino A, Comoglio PM, Crepaldi T. Activation of the MET receptor attenuates doxorubicin-induced cardiotoxicity in vivo and in vitro. Br J Pharmacol 2020; 177:3107-3122. [PMID: 32133617 PMCID: PMC7280013 DOI: 10.1111/bph.15039] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/11/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
Background and Purpose Doxorubicin anti‐cancer therapy is associated with cardiotoxicity, resulting from DNA damage response (DDR). Hepatocyte growth factor (HGF) protects cardiomyocytes from injury, but its effective use is compromised by low biodistribution. In this study, we have investigated whether the activation of the HGF receptor—encoded by the Met gene—by an agonist monoclonal antibody (mAb) could protect against doxorubicin‐induced cardiotoxicity. Experimental Approach The mAb (5 mg·kg−1) was injected in vivo into C57BL/6J mice, before doxorubicin (three doses of 7 mg·kg−1). Cardiac functions were evaluated through MRI after treatment termination. Heart histological staining and mRNA levels of genes associated with heart failure (Acta1 and Nppa), inflammation (IL‐6), and fibrosis (Ctgf, Col1a2, Timp1, and Mmp9) were assessed. MAb (100 nM) was administered in vitro to H9c2 cardiomyoblasts before addition of doxorubicin (25 μM). DDR and apoptosis markers were evaluated by quantitative western blotting, flow cytometry, and immunofluorescence. Stattic was used for pharmacological inactivation of STAT3. Key Results In vivo, administration of the mAb alleviated doxorubicin‐induced cardiac dysfunction and fibrosis. In vitro, mAb mimicked the response to HGF by (a) inhibiting histone H2AX phosphorylation at S139, (b) quenching the expression of the DNA repair enzyme PARP1, and (c) reducing the proteolytic activation of caspase 3. The MET‐driven cardioprotection involved, at least in vitro, the phosphorylation of STAT3. Conclusion and Implications The MET agonist mAb provides a new tool for cardioprotection against anthracycline cardiotoxicity.
Collapse
Affiliation(s)
- Simona Gallo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Martina Spilinga
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| | | | - Giuseppe Ferrauto
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Enza Di Gregorio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Elena Casanova
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | | | | | - Carla Boccaccio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| | - Anna Sapino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Tiziana Crepaldi
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy.,Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|
20
|
Wagner MJ, Khan M, Mohsin S. Healing the Broken Heart; The Immunomodulatory Effects of Stem Cell Therapy. Front Immunol 2020; 11:639. [PMID: 32328072 PMCID: PMC7160320 DOI: 10.3389/fimmu.2020.00639] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/20/2020] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular Disease (CVD) is a leading cause of mortality within the United States. Current treatments being administered to patients who suffered a myocardial infarction (MI) have increased patient survival, but do not facilitate the replacement of damaged myocardium. Recent studies demonstrate that stem cell-based therapies promote myocardial repair; however, the poor engraftment of the transferred stem cell populations within the infarcted myocardium is a major limitation, regardless of the cell type. One explanation for poor cell retention is attributed to the harsh inflammatory response mounted following MI. The inflammatory response coupled to cardiac repair processes is divided into two distinct phases. The first phase is initiated during ischemic injury when necrosed myocardium releases Danger Associated Molecular Patterns (DAMPs) and chemokines/cytokines to induce the activation and recruitment of neutrophils and pro-inflammatory M1 macrophages (MΦs); in turn, facilitating necrotic tissue clearance. During the second phase, a shift from the M1 inflammatory functional phenotype to the M2 anti-inflammatory and pro-reparative functional phenotype, permits the resolution of inflammation and the establishment of tissue repair. T-regulatory cells (Tregs) are also influential in mediating the establishment of the pro-reparative phase by directly regulating M1 to M2 MΦ differentiation. Current studies suggest CD4+ T-lymphocyte populations become activated when presented with autoantigens released from the injured myocardium. The identity of the cardiac autoantigens or paracrine signaling molecules released from the ischemic tissue that directly mediate the phenotypic plasticity of T-lymphocyte populations in the post-MI heart are just beginning to be elucidated. Stem cells are enriched centers that contain a diverse paracrine secretome that can directly regulate responses within neighboring cell populations. Previous studies identify that stem cell mediated paracrine signaling can influence the phenotype and function of immune cell populations in vitro, but how stem cells directly mediate the inflammatory microenvironment of the ischemic heart is poorly characterized and is a topic of extensive investigation. In this review, we summarize the complex literature that details the inflammatory microenvironment of the ischemic heart and provide novel insights regarding how paracrine mediated signaling produced by stem cell-based therapies can regulate immune cell subsets to facilitate pro-reparative myocardial wound healing.
Collapse
Affiliation(s)
- Marcus J Wagner
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Mohsin Khan
- Center for Metabolic Disease, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Physiology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Sadia Mohsin
- Independence Blue Cross Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
21
|
Park JH, Lee NK, Lim HJ, Ji ST, Kim YJ, Jang WB, Kim DY, Kang S, Yun J, Ha JS, Kim H, Lee D, Baek SH, Kwon SM. Pharmacological inhibition of mTOR attenuates replicative cell senescence and improves cellular function via regulating the STAT3-PIM1 axis in human cardiac progenitor cells. Exp Mol Med 2020; 52:615-628. [PMID: 32273566 PMCID: PMC7210934 DOI: 10.1038/s12276-020-0374-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 09/08/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway efficiently regulates the energy state of cells and maintains tissue homeostasis. Dysregulation of the mTOR pathway has been implicated in several human diseases. Rapamycin is a specific inhibitor of mTOR and pharmacological inhibition of mTOR with rapamycin promote cardiac cell generation from the differentiation of mouse and human embryonic stem cells. These studies strongly implicate a role of sustained mTOR activity in the differentiating functions of embryonic stem cells; however, they do not directly address the required effect for sustained mTOR activity in human cardiac progenitor cells. In the present study, we evaluated the effect of mTOR inhibition by rapamycin on the cellular function of human cardiac progenitor cells and discovered that treatment with rapamycin markedly attenuated replicative cell senescence in human cardiac progenitor cells (hCPCs) and promoted their cellular functions. Furthermore, rapamycin not only inhibited mTOR signaling but also influenced signaling pathways, including STAT3 and PIM1, in hCPCs. Therefore, these data reveal a crucial function for rapamycin in senescent hCPCs and provide clinical strategies based on chronic mTOR activity.
Collapse
Affiliation(s)
- Ji Hye Park
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- R&D Center for Advanced Pharmaceuticals & Evaluation, Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, South Korea
| | - Na Kyoung Lee
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Hye Ji Lim
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Seung Taek Ji
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Yeon-Ju Kim
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Woong Bi Jang
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Da Yeon Kim
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Songhwa Kang
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Jisoo Yun
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Jong Seong Ha
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
- Research Institute of Convergence Biomedical Science and Technology, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Hyungtae Kim
- Department of Thoracic and Cardiovascular Surgery, Pusan National University Yangsan Hospital, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea.
| | - Sang Hong Baek
- Laboratory of Cardiovascular Disease, Division of Cardiology, School of Medicine, The Catholic University of Korea, Seoul, 137-040, Republic of Korea.
| | - Sang-Mo Kwon
- Laboratory of Regenerative Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
- Department of Thoracic and Cardiovascular Surgery, Pusan National University Yangsan Hospital, School of Medicine, Pusan National University, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
22
|
Wang J, Chen X, Yang X, Guo B, Li D, Zhu X, Zhang X. Positive role of calcium phosphate ceramics regulated inflammation in the osteogenic differentiation of mesenchymal stem cells. J Biomed Mater Res A 2020; 108:1305-1320. [PMID: 32064734 DOI: 10.1002/jbm.a.36903] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 02/09/2020] [Accepted: 02/12/2020] [Indexed: 02/05/2023]
Abstract
Recently, researches have confirmed the crucial role of inflammatory response in Ca-P ceramic-induced osteogenesis, however, the underlying mechanism has not yet been fully understood. In this study, BCP and β-TCP ceramics were used as material models to investigate the effect of physicochemical properties on inflammatory response in vitro. The results showed that BCP and β-TCP could support macrophages attachment, proliferation, and spreading favorably, as well as promote gene expressions of inflammatory related cytokines (IL-1, IL-6, MCP-1, and TNF-α) and growth factors (TGF-β, FGF, PDGF, VEGF, IGF, and EGF). BCP showed a facilitating function on the gene expressions earlier than β-TCP. Further coculture experiments performed in vitro demonstrated that the CMs containing various increased cytokines for macrophages pre-culture could significantly promote MSCs osteogenic differentiation, which was confirmed by the gene expressions of osteogenic specific markers and the intracellular OCN product accumulation under the stimulation of BCP and β-TCP ceramics. Further evidence was found from the formation of mineralized nodules in BCM and TCM. In addition, this study showed a concise relationship between Ca-P ceramic induced inflammation and its osteoinductivity that the increased cytokines and growth factors from macrophages could promote MSCs osteogenic differentiation.
Collapse
Affiliation(s)
- Jing Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xuening Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xiao Yang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Bo Guo
- Department of Ophthalmology, West China Hospital of Sichuan University, Chengdu, China
| | - Danyang Li
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Nalobin D, Alipkina S, Gaidamaka A, Glukhov A, Khuchua Z. Telomeres and Telomerase in Heart Ontogenesis, Aging and Regeneration. Cells 2020; 9:cells9020503. [PMID: 32098394 PMCID: PMC7072777 DOI: 10.3390/cells9020503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
The main purpose of the review article is to assess the contributions of telomere length and telomerase activity to the cardiac function at different stages of development and clarify their role in cardiac disorders. It has been shown that the telomerase complex and telomeres are of great importance in many periods of ontogenesis due to the regulation of the proliferative capacity of heart cells. The review article also discusses the problems of heart regeneration and the identification of possible causes of dysfunction of telomeres and telomerase.
Collapse
Affiliation(s)
- Denis Nalobin
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
- Correspondence: ; Tel.: +7-916-939-0990
| | - Svetlana Alipkina
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
| | - Anna Gaidamaka
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
| | - Alexander Glukhov
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
- Department of Biochemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russian
| | - Zaza Khuchua
- Department of Biochemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russian
- Institute of Chemical Biology Ilia State University, 0162 Tbilisi, Georgia
- Division of Molecular and Cardiovascular Biology, Cincinnati Children’s Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
24
|
Haider KH, Aramini B. Mircrining the injured heart with stem cell-derived exosomes: an emerging strategy of cell-free therapy. Stem Cell Res Ther 2020; 11:23. [PMID: 31918755 PMCID: PMC6953131 DOI: 10.1186/s13287-019-1548-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/18/2019] [Accepted: 12/29/2019] [Indexed: 02/06/2023] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) have successfully progressed to phase III clinical trials successive to an intensive in vitro and pre-clinical assessment in experimental animal models of ischemic myocardial injury. With scanty evidence regarding their cardiogenic differentiation in the recipient patients' hearts post-engraftment, paracrine secretion of bioactive molecules is being accepted as the most probable underlying mechanism to interpret the beneficial effects of cell therapy. Secretion of small non-coding microRNA (miR) constitutes an integral part of the paracrine activity of stem cells, and there is emerging interest in miRs' delivery to the heart as part of cell-free therapy to exploit their integral role in various cellular processes. MSCs also release membrane vesicles of diverse sizes loaded with a wide array of miRs as part of their paracrine secretions primarily for intercellular communication and to shuttle genetic material. Exosomes can also be loaded with miRs of interest for delivery to the organs of interest including the heart, and hence, exosome-based cell-free therapy is being assessed for cell-free therapy as an alternative to cell-based therapy. This review of literature provides an update on cell-free therapy with primary focus on exosomes derived from BM-derived MSCs for myocardial repair.
Collapse
Affiliation(s)
- Khawaja Husnain Haider
- Sulaiman Alrajhi University, Al-Qaseem, Kingdom of Saudi Arabia
- Department of Basic Sciences, Sulaiman Alrajhi University, PO Box 777, Al Bukairiyah, 51941 Kingdom of Saudi Arabia
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
25
|
Andrade D, Oliveira G, Menezes L, Nascimento AL, Carvalho S, Stumbo AC, Thole A, Garcia-Souza É, Moura A, Carvalho L, Cortez E. Insulin-like growth factor-1 short-period therapy improves cardiomyopathy stimulating cardiac progenitor cells survival in obese mice. Nutr Metab Cardiovasc Dis 2020; 30:151-161. [PMID: 31753790 DOI: 10.1016/j.numecd.2019.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 08/28/2019] [Accepted: 09/02/2019] [Indexed: 01/19/2023]
Abstract
BACKGROUND AND AIMS Cardiovascular diseases are the main cause of mortality in obesity. Despite advanced understanding, the mechanisms that regulate cardiac progenitor cells (CPC) survival in pathological conditions are not clear. Low IGF-1 plasma levels are correlated to obesity, cardiomyopathy and CPC death, so this work aimed to investigate IGF-1 therapeutic potential on cardiomyopathy and its relationship with the survival, proliferation and differentiation of CPC in Western diet-induced obesity. METHODS AND RESULTS Male Swiss mice were divided into control group (CG, n = 8), fed with standard diet; and obese group (OG, n = 16), fed with Western diet, for 12 weeks. At 11th week, OG was subdivided to receive a daily subcutaneous injection of human recombinant IGF-1 (100 μg.Kg-1) for seven consecutive days (OG + IGF1, n = 8). Results showed that IGF-1 therapy improved the metabolic parameters negatively impacted by western diet in OG, reaching levels similar to CG. OG + IGF-1 also demonstrated restored heart energetic metabolism, fibrosis resolution, decreased apoptosis level, restored cardiac gap junctions and intracellular calcium balance. Cardiomyopathy improvement was accompanied by increased CPC survival, proliferation and newly cardiomyocytes formation related to increased pAkt/Akt ratio. CONCLUSION These results suggest that only one week of IGF-1 therapy has cardioprotective effects through Akt pathway upregulation, ensuring CPC survival and differentiation, contributing to heart failure rescue.
Collapse
Affiliation(s)
- Daniela Andrade
- Laboratory of Stem Cell Research, Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, UERJ, Brazil
| | - Genilza Oliveira
- Laboratory of Stem Cell Research, Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, UERJ, Brazil
| | - Luciana Menezes
- Laboratory of Stem Cell Research, Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, UERJ, Brazil
| | - Ana Lúcia Nascimento
- Laboratory of Stem Cell Research, Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, UERJ, Brazil
| | - Simone Carvalho
- Laboratory of Stem Cell Research, Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, UERJ, Brazil
| | - Ana Carolina Stumbo
- Laboratory of Stem Cell Research, Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, UERJ, Brazil
| | - Alessandra Thole
- Laboratory of Stem Cell Research, Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, UERJ, Brazil
| | - Érica Garcia-Souza
- Laboratory of Nutrition Physiology and Development, Department of Physiological Sciences, Institute of Biology, UERJ, Brazil
| | - Anibal Moura
- Laboratory of Nutrition Physiology and Development, Department of Physiological Sciences, Institute of Biology, UERJ, Brazil
| | - Laís Carvalho
- Laboratory of Stem Cell Research, Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, UERJ, Brazil
| | - Erika Cortez
- Laboratory of Stem Cell Research, Department of Histology and Embryology, Institute of Biology, State University of Rio de Janeiro, UERJ, Brazil.
| |
Collapse
|
26
|
Obradovic M, Zafirovic S, Soskic S, Stanimirovic J, Trpkovic A, Jevremovic D, Isenovic ER. Effects of IGF-1 on the Cardiovascular System. Curr Pharm Des 2019; 25:3715-3725. [DOI: 10.2174/1381612825666191106091507] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 10/29/2019] [Indexed: 11/22/2022]
Abstract
:Cardiovascular (CV) diseases are the most common health problems worldwide, with a permanent increase in incidence. Growing evidence underlines that insulin-like growth factor 1 (IGF-1) is a very important hormone responsible for normal CV system physiology. IGF-1 is an anabolic growth hormone, responsible for cell growth, differentiation, proliferation, and survival. Despite systemic effects, IGF-1 exerts a wide array of influences in the CV system affecting metabolic homeostasis, vasorelaxation, cardiac contractility and hypertrophy, autophagy, apoptosis, and antioxidative processes. The vasodilatory effect of IGF-1, is achieved through the regulation of the activity of endothelial nitric oxide synthase (eNOS) and, at least partly, through enhancing inducible NOS (iNOS) activity. Also, IGF-1 stimulates vascular relaxation through regulation of sodium/potassiumadenosine- triphosphatase. Numerous animal studies provided evidence of diverse influences of IGF-1 in the CV system such as vasorelaxation, anti-apoptotic and prosurvival effects. Human studies indicate that low serum levels of free or total IGF-1 contribute to an increased risk of CV and cerebrovascular disease. Large human trials aiming at finding clinical efficacy and outcome of IGF-1-related therapy are of great interest.:We look forward to the development of new IGF 1 therapies with minor side effects. In this review, we discuss the latest literature data regarding the function of IGF-1 in the CV system in the physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Milan Obradovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Sonja Zafirovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Sanja Soskic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Julijana Stanimirovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Andreja Trpkovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| | - Danimir Jevremovic
- Faculty of Stomatology, Pancevo, University Business Academy, 21000 Novi Sad, Serbia
| | - Esma R. Isenovic
- Laboratory of Radiobiology and Molecular Genetics, Vinca Institute of Nuclear Sciences, University of Belgrade, Mike Petrovica Alasa 12-14, 11000 Belgrade, Serbia
| |
Collapse
|
27
|
Ameliorating the Fibrotic Remodeling of the Heart through Direct Cardiac Reprogramming. Cells 2019; 8:cells8070679. [PMID: 31277520 PMCID: PMC6679082 DOI: 10.3390/cells8070679] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Revised: 06/21/2019] [Accepted: 06/23/2019] [Indexed: 12/20/2022] Open
Abstract
Coronary artery disease is the most common form of cardiovascular diseases, resulting in the loss of cardiomyocytes (CM) at the site of ischemic injury. To compensate for the loss of CMs, cardiac fibroblasts quickly respond to injury and initiate cardiac remodeling in an injured heart. In the remodeling process, cardiac fibroblasts proliferate and differentiate into myofibroblasts, which secrete extracellular matrix to support the intact structure of the heart, and eventually differentiate into matrifibrocytes to form chronic scar tissue. Discovery of direct cardiac reprogramming offers a promising therapeutic strategy to prevent/attenuate this pathologic remodeling and replace the cardiac fibrotic scar with myocardium in situ. Since the first discovery in 2010, many progresses have been made to improve the efficiency and efficacy of reprogramming by understanding the mechanisms and signaling pathways that are activated during direct cardiac reprogramming. Here, we overview the development and recent progresses of direct cardiac reprogramming and discuss future directions in order to translate this promising technology into an effective therapeutic paradigm to reverse cardiac pathological remodeling in an injured heart.
Collapse
|
28
|
Mayourian J, Ceholski DK, Gonzalez DM, Cashman TJ, Sahoo S, Hajjar RJ, Costa KD. Physiologic, Pathologic, and Therapeutic Paracrine Modulation of Cardiac Excitation-Contraction Coupling. Circ Res 2019; 122:167-183. [PMID: 29301848 DOI: 10.1161/circresaha.117.311589] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cardiac excitation-contraction coupling (ECC) is the orchestrated process of initial myocyte electrical excitation, which leads to calcium entry, intracellular trafficking, and subsequent sarcomere shortening and myofibrillar contraction. Neurohumoral β-adrenergic signaling is a well-established mediator of ECC; other signaling mechanisms, such as paracrine signaling, have also demonstrated significant impact on ECC but are less well understood. For example, resident heart endothelial cells are well-known physiological paracrine modulators of cardiac myocyte ECC mainly via NO and endothelin-1. Moreover, recent studies have demonstrated other resident noncardiomyocyte heart cells (eg, physiological fibroblasts and pathological myofibroblasts), and even experimental cardiotherapeutic cells (eg, mesenchymal stem cells) are also capable of altering cardiomyocyte ECC through paracrine mechanisms. In this review, we first focus on the paracrine-mediated effects of resident and therapeutic noncardiomyocytes on cardiomyocyte hypertrophy, electrophysiology, and calcium handling, each of which can modulate ECC, and then discuss the current knowledge about key paracrine factors and their underlying mechanisms of action. Next, we provide a case example demonstrating the promise of tissue-engineering approaches to study paracrine effects on tissue-level contractility. More specifically, we present new functional and molecular data on the effects of human adult cardiac fibroblast conditioned media on human engineered cardiac tissue contractility and ion channel gene expression that generally agrees with previous murine studies but also suggests possible species-specific differences. By contrast, paracrine secretions by human dermal fibroblasts had no discernible effect on human engineered cardiac tissue contractile function and gene expression. Finally, we discuss systems biology approaches to help identify key stem cell paracrine mediators of ECC and their associated mechanistic pathways. Such integration of tissue-engineering and systems biology methods shows promise to reveal novel insights into paracrine mediators of ECC and their underlying mechanisms of action, ultimately leading to improved cell-based therapies for patients with heart disease.
Collapse
Affiliation(s)
- Joshua Mayourian
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Delaine K Ceholski
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - David M Gonzalez
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Timothy J Cashman
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Susmita Sahoo
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kevin D Costa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
29
|
Hameed A, Gallagher LB, Dolan E, O’Sullivan J, Ruiz-Hernandez E, Duffy GP, Kelly H. Insulin-like growth factor-1 (IGF-1) poly (lactic-co-glycolic acid) (PLGA) microparticles – development, characterisation, and in vitro assessment of bioactivity for cardiac applications. J Microencapsul 2019; 36:267-277. [DOI: 10.1080/02652048.2019.1622605] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Aamir Hameed
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RSCI), Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland
| | - Laura B. Gallagher
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RSCI), Dublin, Ireland
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Eimear Dolan
- Department of Biomedical Engineering, College of Engineering and Informatics, National University of Ireland Galway, Galway, Ireland
| | - Janice O’Sullivan
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RSCI), Dublin, Ireland
- Department of Anatomy, School of Medicine, College of Medicine, Nursing and Health Science, National University of Ireland Galway, Galway, Ireland
| | - Eduardo Ruiz-Hernandez
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin (TCD), Dublin, Ireland
| | - Garry P. Duffy
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RSCI), Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin (TCD), Dublin, Ireland
- Department of Anatomy, School of Medicine, College of Medicine, Nursing and Health Science, National University of Ireland Galway, Galway, Ireland
- Advanced Materials for Biomedical Engineering and Regenerative Medicine (AMBER), Trinity College Dublin (TCD), Dublin, Ireland
| | - Helena Kelly
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland (RSCI), Dublin, Ireland
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
30
|
Abstract
Non-communicable diseases, such as cardiovascular diseases, are the leading cause of mortality worldwide. For this reason, a tremendous effort is being made worldwide to effectively circumvent these afflictions, where insulin-like growth factor 1 (IGF1) is being proposed both as a marker and as a central cornerstone in these diseases, making it an interesting molecule to focus on. Firstly, at the initiation of metabolic deregulation by overfeeding, IGF1 is decreased/inhibited. Secondly, such deficiency seems to be intimately related to the onset of MetS and establishment of vascular derangements leading to atherosclerosis and finally playing a definitive part in cerebrovascular and myocardial accidents, where IGF1 deficiency seems to render these organs vulnerable to oxidative and apoptotic/necrotic damage. Several human cohort correlations together with basic/translational experimental data seem to confirm deep IGF1 implication, albeit with controversy, which might, in part, be given by experimental design leading to blurred result interpretation.
Collapse
|
31
|
Yousefi M, Mamipour M, Sokullu SE, Ghaderi S, Amini H, Rahbarghazi R. Toll-like receptors in the functional orientation of cardiac progenitor cells. J Cell Physiol 2019; 234:19451-19463. [PMID: 31025370 DOI: 10.1002/jcp.28738] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/04/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
Cardiac progenitor cells (CPCs) have the potential to differentiate into several cell lineages with the ability to restore in cardiac tissue. Multipotency and self-renewal activity are the crucial characteristics of CPCs. Also, CPCs have promising therapeutic roles in cardiac diseases such as valvular disease, thrombosis, atherosclerosis, congestive heart failure, and cardiac remodeling. Toll-like receptors (TLRs), as the main part of the innate immunity, have a key role in the development and differentiation of immune cells. Some reports are found regarding the effect of TLRs in the maturation of stem cells. This article tried to find the potential role of TLRs in the dynamics of CPCs. By showing possible crosstalk between the TLR signaling pathways and CPCs dynamics, we could achieve a better conception related to TLRs in the regeneration of cardiac tissue.
Collapse
Affiliation(s)
- Mohammadreza Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Translational Medicine, Koç University, Istanbul, Turkey
| | - Mina Mamipour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Translational Medicine, Koç University, Istanbul, Turkey
| | - Sadiye E Sokullu
- Engineering Sciences, Bioengineering Department, Faculty of Engineering and Architecture, Izmir Katip Celebi University, Izmir, Turkey
| | - Shahrooz Ghaderi
- Department of System Physiology, Ruhr University, Bochum, Germany
| | - Hassan Amini
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of General and Vascular Surgery, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
32
|
Mohsin S, Houser SR. Cortical Bone Derived Stem Cells for Cardiac Wound Healing. Korean Circ J 2019; 49:314-325. [PMID: 30808081 PMCID: PMC6428954 DOI: 10.4070/kcj.2018.0437] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 12/23/2018] [Indexed: 12/23/2022] Open
Abstract
Ischemic heart disease can lead to myocardial infarction (MI), a major cause of morbidity and mortality worldwide. Adoptive transfer of multiple stem cell types into failing human hearts has demonstrated safety however the beneficial effects in patients with cardiovascular disorders have been modest. Modest improvement in patients with cardiac complications warrants identification of a novel stem cell population that possesses effective reparative properties and improves cardiac function after injury. Recently we have shown in a mouse model and a porcine pre-clinical animal model, that cortical bone derived stem cells (CBSCs) enhance cardiac function after MI and/or ischemia-reperfusion injury. These beneficial effects of allogeneic cell delivery appear to be mediated by paracrine mechanisms rather than by transdifferentiation of injected cells into vessels and/or immature myocytes. This review will discuss role of CBSCs in cardiac wound healing. After having modest beneficial improvement in most of the clinical trials, a critical need is to understand the interaction of the transplanted stem cells with the ischemic cardiac environment. Transplanted stem cells are exposed to pro-inflammatory factors and activated immune cells and fibroblasts, but their interactions remain unknown. We have shown that CBSCs modulate different processes including modulation of the immune response, angiogenesis, and restriction of infarct sizes after cardiac injury. This review will provide information on unique protective signature of CBSCs in rodent/swine animal models for heart repair that should provide basis for developing novel therapies for treating heart failure patients.
Collapse
Affiliation(s)
- Sadia Mohsin
- Department of Pharmacology, Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Steven R. Houser
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
33
|
Conover CA, Bale LK, Frye RL, Schaff HV. Cellular characterization of human epicardial adipose tissue: highly expressed PAPP-A regulates insulin-like growth factor I signaling in human cardiomyocytes. Physiol Rep 2019; 7:e14006. [PMID: 30809969 PMCID: PMC6391584 DOI: 10.14814/phy2.14006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 12/16/2022] Open
Abstract
Little is known about the cellular biology of fat surrounding the human heart. In this study, we obtained paired samples of epicardial fat, the visceral fat depot attached to the heart, and subcutaneous skin fat from patients undergoing open heart surgery to test the hypothesis that human epicardial fat cells differentially express bioactive molecules that have the potential to affect cardiac function. First, we characterized the free fatty acids (FFAs), adipocytokines, and growth factors secreted by isolated adipocytes and preadipocytes in cell culture. There was little to distinguish the fat cell secretory products in terms of FFAs and adipocytokines. The most striking finding was that preadipocytes from epicardial adipose tissue expressed high levels of pregnancy-associated plasma protein-A (PAPP-A), a novel metalloproteinase that enhances local insulin-like growth factor (IGF) action through cleavage of inhibitory IGF binding protein-4 (IGFBP-4). PAPP-A levels were 15-fold higher in conditioned medium from epicardial preadipocytes than from subcutaneous preadipocytes (P < 0.0001). PAPP-A was not expressed in mature adipocytes. Next we determined whether PAPP-A could affect IGF-I signaling in a human cardiomyocyte cell line. IGF-I activated receptor-mediated auto-phosphorylation, and this was blocked by wild-type and protease-resistant IGFBP-4. Addition of PAPP-A induced cleavage of wild-type, but not protease-resistant, IGFBP-4 thereby restoring IGF-I action. A proteolytically defective PAPP-A had no effect. IGF-I receptor-mediated signaling through the phosphatidylinositol 3-kinase pathway was similarly inhibited by IGFBP-4 and restored by PAPP-A. Thus, human epicardial fat cells differentially express PAPP-A, which has the potential to affect IGF signaling in the heart.
Collapse
Affiliation(s)
| | | | - Robert L. Frye
- Department of Cardiovascular DiseasesMayo ClinicRochesterMinnesota
| | | |
Collapse
|
34
|
Wen HJ, Liu GF, Xiao LZ, Wu YG. Involvement of endothelial nitric oxide synthase pathway in IGF‑1 protects endothelial progenitor cells against injury from oxidized LDLs. Mol Med Rep 2018; 19:660-666. [PMID: 30431094 DOI: 10.3892/mmr.2018.9633] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 10/08/2018] [Indexed: 11/05/2022] Open
Abstract
A high level of oxidized low‑density lipoproteins (oxLDLs) is an independent risk factor for cardiovascular disease. The aim of the present study was to investigate whether insulin‑like growth factor‑1 (IGF‑1) protected endothelial progenitor cells (EPCs) from injury caused by ox‑LDLs, and whether the endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) pathway was involved in this process. EPCs were isolated from human peripheral blood and characterized. In order to evaluate the effect of IGF‑1 on EPCs, cells were incubated with ox‑LDLs (100 mg/ml) for 24 h to induce a model of EPC dysfunction in vitro, which demonstrated a decrease in the number of EPCs, concomitant with increased apoptosis and decreased proliferation rates. IGF‑1 dose‑dependently increased the number of EPCs. Concurrently, IGF‑1 decreased the levels of apoptosis of EPCs and improved EPCs proliferation following ox‑LDLs challenge. In addition, IGF‑1 significantly increased NO levels in ox‑LDLs‑treated EPCs, accompanied by an upregulation in eNOS expression. The protective effects of IGF‑1 on EPCs and NO production were abolished by L‑NAME, a specific eNOS inhibitor. These results suggested that IGF‑1 protects EPCs from dysfunction induced by oxLDLs through a mechanism involving the eNOS/NO pathway.
Collapse
Affiliation(s)
- Hao-Jing Wen
- Positron Emission Tomography‑Computed Tomography Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Guo-Feng Liu
- Department of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Li-Zhi Xiao
- Positron Emission Tomography‑Computed Tomography Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yong-Gang Wu
- Positron Emission Tomography‑Computed Tomography Center, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
35
|
Shafiq M, Zhang Y, Zhu D, Zhao Z, Kim DH, Kim SH, Kong D. In situ cardiac regeneration by using neuropeptide substance P and IGF-1C peptide eluting heart patches. Regen Biomater 2018; 5:303-316. [PMID: 30338128 PMCID: PMC6184517 DOI: 10.1093/rb/rby021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 06/26/2018] [Accepted: 07/03/2018] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular diseases cause huge socio-economic burden worldwide. Although a mammalian myocardium has its own limited healing capability, scaffold materials capable of releasing stem cell recruiting/engrafting factors may facilitate the regeneration of the infarcted myocardium. The aim of this research was to develop cardiac patches capable of simultaneously eluting substance P (SP) and insulin-like growth factor-1C (IGF-1C) peptide. Polycaprolactone/collagen type 1-based patches with or without SP and IGF-1C peptide were fabricated by co-electrospinning, which exhibited nanofibrous morphology. SP and IGF-1C/SP patches recruited significantly higher numbers of bone marrow-mesenchymal stem cells than that of the negative control and patch-only groups in vitro. The developed patches were transplanted in an infarcted myocardium for up to 14 days. Mice underwent left anterior descending artery ligation and received one of the following treatments: (i) sham, (ii) saline, (iii) patch-only, (iv) IGF-1C patch, (v) SP patch and (vi) IGF-1C/SP patch. SP and IGF-1C/SP patch-treated groups exhibited better heart function and attenuated adverse cardiac remodeling than that of the saline, patch-only and individual peptide containing cardiac patches. SP patch and IGF-1C/SP patch-treated groups also showed higher numbers of CD31-positive vessels and isolectin B4-positive capillaries than that of other groups. IGF-1C/SP-treated group also showed thicker left ventricular wall in comparison to the saline and patch-only groups. Moreover, IGF-1C/SP patches recruited significantly higher numbers of CD29-positive cells and showed less numbers of Tunel-positive cells compared with the other groups. These data suggest that SP and IGF-1C peptides may act synergistically for in situ tissue repair.
Collapse
Affiliation(s)
- Muhammad Shafiq
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), College of Life Science, Nankai University, Tianjin, China
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul, Republic of Korea
- Department of Chemistry, Center for Tissue Engineering & Regenerative Medicine, Pakistan Institute of Engineering & Applied Sciences (PIEAS), Nilore, Islamabad, Pakistan
| | - Yue Zhang
- Department of Physiology & Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Dashuai Zhu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), College of Life Science, Nankai University, Tianjin, China
| | - Zongxian Zhao
- Department of Physiology & Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Soo Hyun Kim
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, Republic of Korea
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Cheongryang, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), College of Life Science, Nankai University, Tianjin, China
| |
Collapse
|
36
|
Tang J, Wang X, Tan K, Zhu H, Zhang Y, Ouyang W, Liu X, Ding Z. Injury-induced fetal reprogramming imparts multipotency and reparative properties to pericardial adipose stem cells. Stem Cell Res Ther 2018; 9:218. [PMID: 30103817 PMCID: PMC6090634 DOI: 10.1186/s13287-018-0959-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/05/2018] [Accepted: 07/17/2018] [Indexed: 01/15/2023] Open
Abstract
Background Injury may induce a sequential activation of intrinsic reparative activity that supports the maintenance of tissue homeostasis. Method In the present experiments, we investigated whether myocardial infarction (MI) was able to reinstate the expression of Wilms’ tumor factor 1 (WT1) as a key hallmark of fetal reprograming in the pericardial adipose-derived stem cells (pADSC). We characterized the immunophenotypical markers, cardiac potential, and reparative activity of WT1-expressing pADSC (WT1pos) isolated MI Wistar rats with an intact pericardial sac in which cardiac transudate was accumulated, sampled, and analyzed. Results The WT1pos cells formed colony-like aggregates in culture that subsequently generated phase-bright cells that homogenously constituted WT1 expression (> 98%). The WT1pos cells shared identical surface markers with canonical pADSC, but enhanced transcripts for cardiogenesis (isl-1, gata-4, Sox2 and Tbx18) as well as cardiac commitment (endothelial: 28%; cardiomyogenic: 12.3%) in defined conditions. Remarkably, cardiac transplantation of WT1pos cells promoted regional angiogenesis and myogenesis which led to significant functional amelioration of the infarcted hearts. Furthermore, we demonstrated that WT1pos cells uniquely secreted hepatocyte growth factor (HGF) as a key antiapoptotic factor that promotes cardiac repair. Conclusion Injury-associated fetal reprogramming in pADSC facilitates cardiac differentiation and promotes the reparative activity by enhancing HGF production. As such, injury-“conditioned” pADSC may represent a useful autologous cell donor from infarcted patients for cell-based therapy.
Collapse
Affiliation(s)
- Jianfeng Tang
- Department of Cardiology, Danyang People's Hospital, West Xinmin Rd. 5, 212300, Danyang, People's Republic of China
| | - Xiaoming Wang
- Department of Cardiothoracic Surgery, BenQ Medical Center, Hexi Rd. 17, 210019, Nanjing, People's Republic of China
| | - Kezhe Tan
- Department of Anesthesiology, Changhai Hospital, Naval Medical University, Changhai Rd. 168, 200433, Shanghai, People's Republic of China
| | - Hongtao Zhu
- Department of Cardiology, Danyang People's Hospital, West Xinmin Rd. 5, 212300, Danyang, People's Republic of China
| | - Youming Zhang
- Department of Cardiology, Danyang People's Hospital, West Xinmin Rd. 5, 212300, Danyang, People's Republic of China
| | - Weili Ouyang
- Department of Cardiology, Danyang People's Hospital, West Xinmin Rd. 5, 212300, Danyang, People's Republic of China
| | - Xueqing Liu
- Department of Cardiology, Danyang People's Hospital, West Xinmin Rd. 5, 212300, Danyang, People's Republic of China.
| | - Zhaoping Ding
- Department of Cardiology, Danyang People's Hospital, West Xinmin Rd. 5, 212300, Danyang, People's Republic of China. .,Department of Anesthesiology, Changhai Hospital, Naval Medical University, Changhai Rd. 168, 200433, Shanghai, People's Republic of China. .,Institue of Molecular Cardiology, Heinrich-Heine University of Düsseldorf, Moorenstr. 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
37
|
Cianflone E, Aquila I, Scalise M, Marotta P, Torella M, Nadal-Ginard B, Torella D. Molecular basis of functional myogenic specification of Bona Fide multipotent adult cardiac stem cells. Cell Cycle 2018; 17:927-946. [PMID: 29862928 PMCID: PMC6103696 DOI: 10.1080/15384101.2018.1464852] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 06/01/2018] [Accepted: 04/08/2018] [Indexed: 01/14/2023] Open
Abstract
Ischemic Heart Disease (IHD) remains the developed world's number one killer. The improved survival from Acute Myocardial Infarction (AMI) and the progressive aging of western population brought to an increased incidence of chronic Heart Failure (HF), which assumed epidemic proportions nowadays. Except for heart transplantation, all treatments for HF should be considered palliative because none of the current therapies can reverse myocardial degeneration responsible for HF syndrome. To stop the HF epidemic will ultimately require protocols to reduce the progressive cardiomyocyte (CM) loss and to foster their regeneration. It is now generally accepted that mammalian CMs renew throughout life. However, this endogenous regenerative reservoir is insufficient to repair the extensive damage produced by AMI/IHD while the source and degree of CM turnover remains strongly disputed. Independent groups have convincingly shown that the adult myocardium harbors bona-fide tissue specific cardiac stem cells (CSCs). Unfortunately, recent reports have challenged the identity and the endogenous myogenic capacity of the c-kit expressing CSCs. This has hampered progress and unless this conflict is settled, clinical tests of repair/regenerative protocols are unlikely to provide convincing answers about their clinical potential. Here we review recent data that have eventually clarified the specific phenotypic identity of true multipotent CSCs. These cells when coaxed by embryonic cardiac morphogens undergo a precisely orchestrated myogenic commitment process robustly generating bona-fide functional cardiomyocytes. These data should set the path for the revival of further investigation untangling the regenerative biology of adult CSCs to harness their potential for HF prevention and treatment.
Collapse
Affiliation(s)
- Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Iolanda Aquila
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Pina Marotta
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Michele Torella
- Department of Cardiothoracic Sciences, University of Campania Campus “Salvatore Venuta” Viale Europa- Loc. Germaneto “L. Vanvitelli”, Naples, Italy
| | - Bernardo Nadal-Ginard
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
38
|
Sustained release of targeted cardiac therapy with a replenishable implanted epicardial reservoir. Nat Biomed Eng 2018; 2:416-428. [DOI: 10.1038/s41551-018-0247-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 05/09/2018] [Indexed: 12/12/2022]
|
39
|
Vigneault P, Naud P, Qi X, Xiao J, Villeneuve L, Davis DR, Nattel S. Calcium-dependent potassium channels control proliferation of cardiac progenitor cells and bone marrow-derived mesenchymal stem cells. J Physiol 2018; 596:2359-2379. [PMID: 29574723 DOI: 10.1113/jp275388] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 02/26/2018] [Indexed: 12/26/2022] Open
Abstract
KEY POINTS Ex vivo proliferated c-Kit+ endogenous cardiac progenitor cells (eCPCs) obtained from mouse and human cardiac tissues have been reported to express a wide range of functional ion channels. In contrast to previous reports in cultured c-Kit+ eCPCs, we found that ion currents were minimal in freshly isolated cells. However, inclusion of free Ca2+ intracellularly revealed a prominent inwardly rectifying current identified as the intermediate conductance Ca2+ -activated K+ current (KCa3.1) Electrical function of both c-Kit+ eCPCs and bone marrow-derived mesenchymal stem cells is critically governed by KCa3.1 calcium-dependent potassium channels. Ca2+ -induced increases in KCa3.1 conductance are necessary to optimize membrane potential during Ca2+ entry. Membrane hyperpolarization due to KCa3.1 activation maintains the driving force for Ca2+ entry that activates stem cell proliferation. Cardiac disease downregulates KCa3.1 channels in resident cardiac progenitor cells. Alterations in KCa3.1 may have pathophysiological and therapeutic significance in regenerative medicine. ABSTRACT Endogenous c-Kit+ cardiac progenitor cells (eCPCs) and bone marrow (BM)-derived mesenchymal stem cells (MSCs) are being developed for cardiac regenerative therapy, but a better understanding of their physiology is needed. Here, we addressed the unknown functional role of ion channels in freshly isolated eCPCs and expanded BM-MSCs using patch-clamp, microfluorometry and confocal microscopy. Isolated c-Kit+ eCPCs were purified from dog hearts by immunomagnetic selection. Ion currents were barely detectable in freshly isolated c-Kit+ eCPCs with buffering of intracellular calcium (Ca2+i ). Under conditions allowing free intracellular Ca2+ , freshly isolated c-Kit+ eCPCs and ex vivo proliferated BM-MSCs showed prominent voltage-independent conductances that were sensitive to intermediate-conductance K+ -channel (KCa3.1 current, IKCa3.1 ) blockers and corresponding gene (KCNN4)-expression knockdown. Depletion of Ca2+i induced membrane-potential (Vmem ) depolarization, while store-operated Ca2+ entry (SOCE) hyperpolarized Vmem in both cell types. The hyperpolarizing SOCE effect was substantially reduced by IKCa3.1 or SOCE blockade (TRAM-34, 2-APB), and IKCa3.1 blockade (TRAM-34) or KCNN4-knockdown decreased the Ca2+ entry resulting from SOCE. IKCa3.1 suppression reduced c-Kit+ eCPC and BM-MSC proliferation, while significantly altering the profile of cyclin expression. IKCa3.1 was reduced in c-Kit+ eCPCs isolated from dogs with congestive heart failure (CHF), along with corresponding KCNN4 mRNA. Under perforated-patch conditions to maintain physiological [Ca2+ ]i , c-Kit+ eCPCs from CHF dogs had less negative resting membrane potentials (-58 ± 7 mV) versus c-Kit+ eCPCs from control dogs (-73 ± 3 mV, P < 0.05), along with slower proliferation. Our study suggests that Ca2+ -induced increases in IKCa3.1 are necessary to optimize membrane potential during the Ca2+ entry that activates progenitor cell proliferation, and that alterations in KCa3.1 may have pathophysiological and therapeutic significance in regenerative medicine.
Collapse
Affiliation(s)
- Patrick Vigneault
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Patrice Naud
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Xiaoyan Qi
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Jiening Xiao
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Louis Villeneuve
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada
| | - Darryl R Davis
- University of Ottawa Heart Institute, Ottawa, Ontario, Canada
| | - Stanley Nattel
- Research Center and Department of Medicine, Montreal Heart Institute, Université de Montréal, Montreal, Quebec, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.,Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
40
|
Chen W, Ju J, Yang Y, Wang H, Chen W, Zhao X, Ye H, Zhang Y. Astragalus polysaccharides protect cardiac stem and progenitor cells by the inhibition of oxidative stress-mediated apoptosis in diabetic hearts. DRUG DESIGN DEVELOPMENT AND THERAPY 2018; 12:943-954. [PMID: 29719380 PMCID: PMC5916262 DOI: 10.2147/dddt.s155686] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction Diabetic cardiomyopathy is characterized by an imbalance between myocyte death and regeneration mediated by the progressive loss of cardiac stem and progenitor cells (CSPCs) by apoptosis and necrosis due to the activation of oxidative stress with diabetes. In this study, we evaluated the beneficial effect of astragalus polysaccharides (APS) therapy on the protection of CSPCs through its antioxidative capacity in diabetic hearts. Materials and methods Streptozotocin (STZ)-induced diabetic mice and heterozygous (SOD2+/−) knockout mice were employed and administered with APS. Ventricular CSPCs were isolated for oxidative evaluation. The abundance, apoptosis and proliferation, reactive oxygen species (ROS) formation, oxidative damage, and SOD2 protein levels and activities were evaluated in ventricular CSPCs. Results We confirmed that APS increased the CSPC abundance, reduced the apoptosis of CSPCs, and enhanced the proliferation of CSPCs in both STZ-induced diabetic mice and nondiabetic SOD2+/− mice. In addition, therapy of APS enhanced SOD2 protein levels and enzyme activities, and inhibited ROS formation and oxidative damage of CSPCs from both STZ-induced diabetic mice and nondiabetic SOD2+/− mice. Conclusion Our findings demonstrated the positive effect of APS on the rescue of CSPC preservation in diabetes, dependent on the inhibition of oxidative stress-mediated apoptosis.
Collapse
Affiliation(s)
- Wei Chen
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, China
| | - Jing Ju
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, China
| | - Yehong Yang
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Core Center of Animal Facility, School of Medicine, Fudan University, Shanghai, China
| | - Wenjie Chen
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, China
| | - Xuelan Zhao
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongying Ye
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yu Zhang
- Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Rizzo P, Bollini S, Bertero E, Ferrari R, Ameri P. Beyond cardiomyocyte loss: Role of Notch in cardiac aging. J Cell Physiol 2018; 233:5670-5683. [PMID: 29271542 DOI: 10.1002/jcp.26417] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 12/05/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022]
Abstract
The knowledge of the cellular events occurring in the aging heart has dramatically expanded in the last decade and is expected to further grow in years to come. It is now clear that impaired function and loss of cardiomyocytes are major features of cardiac aging, but other events are likewise important. In particular, accumulating experimental evidence highlights the importance of fibroblast and cardiac progenitor cell (CPC) dysfunction. The Notch pathway regulates cardiomyocyte, fibroblast, and CPC activity and, thus, may be critically involved in heart disease associated with advanced age, especially heart failure. In a translational perspective, thorough investigation of the Notch system in the aging myocardium may lead to the identification of molecular targets for novel therapies for age-related cardiac disease.
Collapse
Affiliation(s)
- Paola Rizzo
- Department of Morphology, Surgery, and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.,Maria Cecilia Hospital, GVM Care and Research, E.S. Health Science Foundation, Cotignola, Italy
| | - Sveva Bollini
- Department of Experimental Medicine, Regenerative Medicine Laboratory, University of Genova, Genova, Italy
| | - Edoardo Bertero
- Department of Internal Medicine, Laboratory of Cardiovascular Biology, University of Genova and Ospedale Policlinico San Martino IRCCS per Oncologia, Genova, Italy
| | - Roberto Ferrari
- Maria Cecilia Hospital, GVM Care and Research, E.S. Health Science Foundation, Cotignola, Italy
| | - Pietro Ameri
- Department of Internal Medicine, Laboratory of Cardiovascular Biology, University of Genova and Ospedale Policlinico San Martino IRCCS per Oncologia, Genova, Italy
| |
Collapse
|
42
|
Marotta P, Cianflone E, Aquila I, Vicinanza C, Scalise M, Marino F, Mancuso T, Torella M, Indolfi C, Torella D. Combining cell and gene therapy to advance cardiac regeneration. Expert Opin Biol Ther 2018; 18:409-423. [PMID: 29347847 DOI: 10.1080/14712598.2018.1430762] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The characterization of multipotent endogenous cardiac stem cells (eCSCs) and the breakthroughs of somatic cell reprogramming to boost cardiomyocyte replacement have fostered the prospect of achieving functional heart repair/regeneration. AREAS COVERED Allogeneic CSC therapy through its paracrine stimulation of the endogenous resident reparative/regenerative process produces functional meaningful myocardial regeneration in pre-clinical porcine myocardial infarction models and is currently tested in the first-in-man human trial. The in vivo test of somatic reprogramming and cardioregenerative non-coding RNAs revived the interest in gene therapy for myocardial regeneration. The latter, together with the advent of genome editing, has prompted most recent efforts to produce genetically-modified allogeneic CSCs that secrete cardioregenerative factors to optimize effective myocardial repair. EXPERT OPINION The current war against heart failure epidemics in western countries seeks to find effective treatments to set back the failing hearts prolonging human lifespan. Off-the-shelf allogeneic-genetically-modified CSCs producing regenerative agents are a novel and evolving therapy set to be affordable, safe, effective and available at all times for myocardial regeneration to either prevent or treat heart failure.
Collapse
Affiliation(s)
- Pina Marotta
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Eleonora Cianflone
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Iolanda Aquila
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Carla Vicinanza
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Mariangela Scalise
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Fabiola Marino
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Teresa Mancuso
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Michele Torella
- b Department of Cardiothoracic Sciences , University of Campania "L. Vanvitelli" , Naples , Italy
| | - Ciro Indolfi
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| | - Daniele Torella
- a Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences , Magna Graecia University , Catanzaro , Italy
| |
Collapse
|
43
|
Renko O, Tolonen AM, Rysä J, Magga J, Mustonen E, Ruskoaho H, Serpi R. SDF1 gradient associates with the distribution of c-Kit+ cardiac cells in the heart. Sci Rep 2018; 8:1160. [PMID: 29348441 PMCID: PMC5773575 DOI: 10.1038/s41598-018-19417-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 12/29/2017] [Indexed: 12/11/2022] Open
Abstract
Identification of the adult cardiac stem cells (CSCs) has offered new therapeutic possibilities for treating ischemic myocardium. CSCs positive for the cell surface antigen c-Kit are known as the primary source for cardiac regeneration. Accumulating evidence shows that chemokines play important roles in stem cell homing. Here we investigated molecular targets to be utilized in modulating the mobility of endogenous CSCs. In a four week follow-up after experimental acute myocardial infarction (AMI) with ligation of the left anterior descending (LAD) coronary artery of Sprague-Dawley rats c-Kit+ CSCs redistributed in the heart. The number of c-Kit+ CSCs in the atrial c-Kit niche was diminished, whereas increased amount was observed in the left ventricle and apex. This was associated with increased expression of stromal cell-derived factor 1 alpha (SDF1α), and a significant positive correlation was found between c-Kit+ CSCs and SDF1α expression in the heart. Moreover, the migratory capacity of isolated c-Kit+ CSCs was induced by SDF1 treatment in vitro. We conclude that upregulation of SDF1α after AMI associates with increased expression of endogenous c-Kit+ CSCs in the injury area, and show induced migration of c-Kit+ cells by SDF1.
Collapse
Affiliation(s)
- Outi Renko
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Anna-Maria Tolonen
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Jaana Rysä
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Johanna Magga
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Erja Mustonen
- Research Unit of Biomedicine, Department of Pharmacology and Toxicology, University of Oulu, Oulu, Finland
| | - Heikki Ruskoaho
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Raisa Serpi
- Biocenter Oulu, Faculty of Biochemistry and Molecular Medicine, Oulu Center for Cell-Matrix Research, University of Oulu, Oulu, Finland.
| |
Collapse
|
44
|
Cappetta D, Rossi F, Piegari E, Quaini F, Berrino L, Urbanek K, De Angelis A. Doxorubicin targets multiple players: A new view of an old problem. Pharmacol Res 2018; 127:4-14. [DOI: 10.1016/j.phrs.2017.03.016] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 01/22/2023]
|
45
|
Cellular Therapeutics for Heart Failure: Focus on Mesenchymal Stem Cells. Stem Cells Int 2017; 2017:9640108. [PMID: 29391871 PMCID: PMC5748110 DOI: 10.1155/2017/9640108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 07/31/2017] [Accepted: 08/14/2017] [Indexed: 12/28/2022] Open
Abstract
Resulting from a various etiologies, the most notable remains ischemia; heart failure (HF) manifests as the common end pathway of many cardiovascular processes and remains among the top causes for hospitalization and a major cause of morbidity and mortality worldwide. Current pharmacologic treatment for HF utilizes pharmacologic agents to control symptoms and slow further deterioration; however, on a cellular level, in a patient with progressive disease, fibrosis and cardiac remodeling can continue leading to end-stage heart failure. Cellular therapeutics have risen as the new hope for an improvement in the treatment of HF. Mesenchymal stem cells (MSCs) have gained popularity given their propensity of promoting endogenous cellular repair of a myriad of disease processes via paracrine signaling through expression of various cytokines, chemokines, and adhesion molecules resulting in activation of signal transduction pathways. While the exact mechanism remains to be completely elucidated, this remains the primary mechanism identified to date. Recently, MSCs have been incorporated as the central focus in clinical trials investigating the role how MSCs can play in the treatment of HF. In this review, we focus on the characteristics of MSCs that give them a distinct edge as cellular therapeutics and present results of clinical trials investigating MSCs in the setting of ischemic HF.
Collapse
|
46
|
Madonna R, Cevik C, Nasser M, De Caterina R. Hepatocyte growth factor: Molecular biomarker and player in cardioprotection and cardiovascular regeneration. Thromb Haemost 2017; 107:656-61. [DOI: 10.1160/th11-10-0711] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 01/03/2012] [Indexed: 11/05/2022]
Abstract
SummaryThe liver possesses impressive regenerative capacities. Grafts of embryonic liver explants and liver explant-conditioned media have been shown to enhance the mitotic activity of hepatocytes. Hepatocyte growth factor (HGF), also named scatter factor (SF), has been identified as a primary candidate in promoting and regulating liver regeneration. Although initially thought to be a liver-specific mitogen, HGF was later reported to have mitogenic, motogenic, morphogenic, and anti-apoptotic activities in various cell types. By promoting angiogenesis and inhibiting apoptosis, endogenous HGF may play an important role in cardioprotection as well as in the regeneration of endothelial cells and cardiomyocytes after myocardial infarction. Since serum concentration of HGF increases in the early phase of myocardial infarction and in heart failure, HGF may also play a key role as a prognostic and diagnostic biomarker of cardiovascular disease. Here we discuss the role of HGF as a biomarker and mediator in cardioprotection and cardiovascular regeneration.
Collapse
|
47
|
Ling L, Gu S, Cheng Y, Ding L. bFGF promotes Sca‑1+ cardiac stem cell migration through activation of the PI3K/Akt pathway. Mol Med Rep 2017; 17:2349-2356. [PMID: 29207135 PMCID: PMC5783475 DOI: 10.3892/mmr.2017.8178] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/21/2016] [Indexed: 01/19/2023] Open
Abstract
Cardiac stem cells (CSCs) are important for improving cardiac function following myocardial infarction, with CSC migration to infarcted or ischemic myocardium important for cardiac regeneration. Strategies to improve cell migration may improve the efficiency of myocardial regeneration. Basic fibroblast growth factor (bFGF) is an essential molecule in cell migration, but the endogenous bFGF level is too low to be effective. The effect of exogenously delivered bFGF on CSC migration was observed in vitro and in vivo in the present study. The CSC migration index in response to various bFGF concentrations was demonstrated in vitro. In addition, a murine myocardial infarction model was constructed and bFGF protein expression levels and CSC aggregation following myocardial infarction were observed. To study cell migration in vivo, CM-Dil-labeled CSCs or bFGF-CSCs were injected into the peri-infarct myocardium following myocardium infarction and cell migration and maintenance in the peri-infarct/infarct area was observed 1 week later. Protein expression levels of bFGF, CXCR-4 and SDF-1 were assessed, as was myocardium capillary density. The Akt inhibitor deguelin was used to assess the role of the PI3K/Akt pathway in vitro and in vivo. The present study demonstrated that bFGF-promoted Sca-1+ CSC migration, with the highest migration rate occurring at a concentration of 45 ng/ml. The PI3K/Akt pathway inhibitor deguelin attenuated this increase. The phospho-Akt/Akt ratio was elevated significantly after 30 min of bFGF exposure. Transplantation of bFGF-treated Sca-1+ CSCs led to improved cell maintenance in the peri-infarct area and increased cell migration to the infarct area, as well as improved angiogenesis. Protein expression levels of bFGF, CXCR-4 and SDF-1 were upregulated, and this upregulation was partially attenuated by deguelin. Therefore, bFGF was demonstrated to promote Sca-1+ CSC migration both in vitro and in vivo, partially through activation of the PI3K/Akt pathway. This may provide a new method for facilitating CSC therapy for myocardium repair after myocardium injury.
Collapse
Affiliation(s)
- Lin Ling
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Shaohua Gu
- Department of Nephrology, The Third People's Hospital of Kunshan, Kunshan, Jiangsu 215300, P.R. China
| | - Yan Cheng
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214000, P.R. China
| | - Liucheng Ding
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, P.R. China
| |
Collapse
|
48
|
Detert S, Stamm C, Beez C, Diedrichs F, Ringe J, Van Linthout S, Seifert M, Tschöpe C, Sittinger M, Haag M. The atrial appendage as a suitable source to generate cardiac-derived adherent proliferating cells for regenerative cell-based therapies. J Tissue Eng Regen Med 2017; 12:e1404-e1417. [PMID: 28752609 DOI: 10.1002/term.2528] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 06/22/2017] [Accepted: 07/24/2017] [Indexed: 12/20/2022]
Abstract
Cardiac-derived adherent proliferating (CardAP) cells obtained from endomyocardial biopsies (EMBs) with known anti-fibrotic and pro-angiogenic properties are good candidates for the autologous therapy of end-stage cardiac diseases such as dilated cardiomyopathy. However, due to the limited number of CardAP cells that can be obtained from EMBs, our aim is to isolate cells with similar properties from other regions of the heart with comparable tissue architecture. Here, we introduce the atrial appendage as a candidate region. Atrial appendage-derived cells were sorted with CD90 microbeads to obtain a CD90low cell population, which were subsequently analysed for their surface marker and gene expression profiles via flow cytometry and micro array analysis. Enzyme-linked immunosorbent assays for vascular endothelial growth factor and interleukin-8 as well as tube formation assays were performed to investigate pro-angiogenic properties. Furthermore, growth kinetic assays were performed to estimate the cell numbers needed for cell-based products. Microarray analysis revealed the expression of numerous pro-angiogenic genes and strong similarities to CardAP cells with which they also share expression levels of defined surface antigens, that is, CD29+ , CD44+ , CD45- , CD73+ , CD90low , CD105+ , and CD166+ . High secretion levels of vascular endothelial growth factor and interleukin-8 as well as improved properties of vascular structures in vitro could be detected. Based on growth parameters, cell dosages for the treatment of more than 250 patients are possible using one appendage. These results lead to the conclusion that isolating cells with regenerative characteristics from atrial appendages is feasible and permits further investigations towards allogenic cell-based therapies.
Collapse
Affiliation(s)
- Stephan Detert
- Tissue Engineering Laboratory, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | - Christien Beez
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Falk Diedrichs
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Jochen Ringe
- Tissue Engineering Laboratory, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sophie Van Linthout
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Martina Seifert
- Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Carsten Tschöpe
- Department of Cardiology, Charité-Universitätsmedizin Berlin, Berlin, Germany.,DZHK (German Center for Cardiovascular Research), partner site Berlin, Germany
| | - Michael Sittinger
- Tissue Engineering Laboratory, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marion Haag
- Tissue Engineering Laboratory, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
49
|
Roura S, Gálvez-Montón C, Mirabel C, Vives J, Bayes-Genis A. Mesenchymal stem cells for cardiac repair: are the actors ready for the clinical scenario? Stem Cell Res Ther 2017; 8:238. [PMID: 29078809 PMCID: PMC5658929 DOI: 10.1186/s13287-017-0695-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
For years, sufficient progress has been made in treating heart failure following myocardial infarction; however, the social and economic burdens and the costs to world health systems remain high. Moreover, treatment advances have not resolved the underlying problem of functional heart tissue loss. In this field of research, for years we have actively explored innovative biotherapies for cardiac repair. Here, we present a general, critical overview of our experience in using mesenchymal stem cells, derived from cardiac adipose tissue and umbilical cord blood, in a variety of cell therapy and tissue engineering approaches. We also include the latest advances and future challenges, including good manufacturing practice and regulatory issues. Finally, we evaluate whether recent approaches hold potential for reliable translation to clinical trials.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain. .,Center of Regenerative Medicine in Barcelona, Barcelona, Spain. .,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain. .,ICREC (Heart Failure and Cardiac Regeneration) Research Programme, Health Sciences Research Institute Germans Trias i Pujol (IGTP), Carretera de Can Ruti, Camí de les Escoles s/n, 08916, Badalona, Barcelona, Spain.
| | - Carolina Gálvez-Montón
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain.,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain
| | - Clémentine Mirabel
- Servei de Teràpia Cel∙lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain
| | - Joaquim Vives
- Servei de Teràpia Cel∙lular, Banc de Sang i Teixits, Edifici Dr. Frederic Duran i Jordà, Passeig Taulat, 116, 08005, Barcelona, Spain.,Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Passeig de la Vall d'Hebron 129-139, 08035, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Antoni Bayes-Genis
- ICREC Research Program, Germans Trias i Pujol Health Research Institute, Badalona, Spain. .,CIBERCV, Instituto de Salud Carlos III, Madrid, Spain. .,Department of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain. .,Cardiology Service, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain. .,Heart Institute, Hospital Universitari Germans Trias i Pujol University Hospital, Carretera de Canyet s/n, 08916, Badalona, Barcelona, Spain.
| |
Collapse
|
50
|
Cardiac Nonmyocyte Cell Functions and Crosstalks in Response to Cardiotoxic Drugs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1089359. [PMID: 29201269 PMCID: PMC5671742 DOI: 10.1155/2017/1089359] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/24/2017] [Indexed: 01/06/2023]
Abstract
The discovery of the molecular mechanisms involved in the cardiac responses to anticancer drugs represents the current goal of cardio-oncology research. The oxidative stress has a pivotal role in cardiotoxic responses, affecting the function of all types of cardiac cells, and their functional crosstalks. Generally, cardiomyocytes are the main target of research studies on cardiotoxicity, but recently the contribution of the other nonmyocyte cardiac cells is becoming of growing interest. This review deals with the role of oxidative stress, induced by anticancer drugs, in cardiac nonmyocyte cells (fibroblasts, vascular cells, and immune cells). The alterations of functional interplays among these cardiac cells are discussed, as well. These interesting recent findings increase the knowledge about cardiotoxicity and suggest new molecular targets for both diagnosis and therapy.
Collapse
|