1
|
Bibi A, Madè A, Greco S, Garcia-Manteiga JM, Tascini AS, Tastsoglou S, Zaccagnini G, Leszek P, Gaetano C, Martelli F. Circular PVT1 promotes cardiac fibroblast activation interacting with miR-30a-5p and miR-125b-5p. Cell Death Dis 2025; 16:325. [PMID: 40258819 PMCID: PMC12012019 DOI: 10.1038/s41419-025-07652-7] [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: 06/07/2024] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/23/2025]
Abstract
Circular RNAs (circRNAs) are involved in the pathogenesis of several cardiovascular diseases, including heart failure. In this study, we report that circular PVT1 (circPVT1) was upregulated in the left ventricle of 31 ischemic heart failure patients compared to 11 non-ischemic controls. RNA sequencing analysis following circPVT1 knockdown in immortalized human cardiomyocytes identified differentially expressed genes, mainly involved in fibrosis. Notably, in human cardiac fibroblasts, circPVT1 expression significantly increased after TGF-β1 treatment and circPVT1 silencing attenuated the levels of pro-fibrotic markers induced by TGF-β1. RNA pull-down assays validated the interaction between circPVT1 and two fibrosis-related miRNAs, miR-30a-5p and miR-125b-5p. The levels of these miRNAs were not altered upon circPVT1 knockdown. However, the expression of their mRNA targets was deregulated upon circPVT1 silencing, suggesting that circPVT1 modulates miRNA cellular bioavailability. Accordingly, inhibition of either miR-30a-5p or miR-125b-5p restored the expression of TGF-β1-induced pro-fibrotic markers following circPVT1 silencing, indicating that both miR-30a-5p and miR-125b-5p act as downstream effectors of circPVT1 in cardiac fibroblast activation. In conclusion, these findings highlight a pro-fibrotic role for circPVT1, which can regulate cardiac fibroblast activation interacting with the anti-fibrotic miR-30a-5p and miR-125b-5p. The modulation of circPVT1 expression may represent a potential strategy to reduce cardiac fibrosis and remodeling.
Collapse
Affiliation(s)
- Alessia Bibi
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Alisia Madè
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | | | - Anna Sofia Tascini
- Center for Omics Sciences, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Spyros Tastsoglou
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Germana Zaccagnini
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, Department of Mechanical Circulatory Support and Transplant, National Institute of Cardiology, Warsaw, Poland
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, Milan, Italy.
- Laboratory of Stem Cell Biology, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania.
| |
Collapse
|
2
|
Berezin AE, Berezin AA. Extracellular vesicles in heart failure. Adv Clin Chem 2024; 119:1-32. [PMID: 38514208 DOI: 10.1016/bs.acc.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Physiologically, extracellular vesicles (EVs) have been implicated as crucial mediators of immune response, cell homeostasis, angiogenesis, cell differentiation and growth, and tissue repair. In heart failure (HF) they may act as regulators of cardiac remodeling, microvascular inflammation, micro environmental changes, tissue fibrosis, atherosclerosis, neovascularization of plaques, endothelial dysfunction, thrombosis, and reciprocal heart-remote organ interaction. The chapter summaries the nomenclature, isolation, detection of EVs, their biologic role and function physiologically as well as in the pathogenesis of HF. Current challenges to the utilization of EVs as diagnostic and predictive biomarkers in HF are also discussed.
Collapse
Affiliation(s)
- Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, Salzburg, Austria.
| | | |
Collapse
|
3
|
Lin LC, Liu ZY, Tu B, Song K, Sun H, Zhou Y, Sha JM, Zhang Y, Yang JJ, Zhao JY, Tao H. Epigenetic signatures in cardiac fibrosis: Focusing on noncoding RNA regulators as the gatekeepers of cardiac fibroblast identity. Int J Biol Macromol 2024; 254:127593. [PMID: 37898244 DOI: 10.1016/j.ijbiomac.2023.127593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 09/13/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023]
Abstract
Cardiac fibroblasts play a pivotal role in cardiac fibrosis by transformation of fibroblasts into myofibroblasts, which synthesis and secrete a large number of extracellular matrix proteins. Ultimately, this will lead to cardiac wall stiffness and impaired cardiac performance. The epigenetic regulation and fate reprogramming of cardiac fibroblasts has been advanced considerably in recent decades. Non coding RNAs (microRNAs, lncRNAs, circRNAs) regulate the functions and behaviors of cardiac fibroblasts, including proliferation, migration, phenotypic transformation, inflammation, pyroptosis, apoptosis, autophagy, which can provide the basis for novel targeted therapeutic treatments that abrogate activation and inflammation of cardiac fibroblasts, induce different death pathways in cardiac fibroblasts, or make it sensitive to established pathogenic cells targeted cytotoxic agents and biotherapy. This review summarizes our current knowledge in this field of ncRNAs function in epigenetic regulation and fate determination of cardiac fibroblasts as well as the details of signaling pathways contribute to cardiac fibrosis. Moreover, we will comment on the emerging landscape of lncRNAs and circRNAs function in regulating signal transduction pathways, gene translation processes and post-translational regulation of gene expression in cardiac fibroblast. In the end, the prospect of cardiac fibroblasts targeted therapy for cardiac fibrosis based on ncRNAs is discussed.
Collapse
Affiliation(s)
- Li-Chan Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Zhi-Yan Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Bin Tu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Kai Song
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - He Sun
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Yang Zhou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Ji-Ming Sha
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China
| | - Ye Zhang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China.
| | - Jian-Yuan Zhao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, PR China; Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, PR China.
| |
Collapse
|
4
|
Zhang H, Zhou Y, Wen D, Wang J. Noncoding RNAs: Master Regulator of Fibroblast to Myofibroblast Transition in Fibrosis. Int J Mol Sci 2023; 24:1801. [PMID: 36675315 PMCID: PMC9861037 DOI: 10.3390/ijms24021801] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Myofibroblasts escape apoptosis and proliferate abnormally under pathological conditions, especially fibrosis; they synthesize and secrete a large amount of extracellular matrix (ECM), such as α-SMA and collagen, which leads to the distortion of organ parenchyma structure, an imbalance in collagen deposition and degradation, and the replacement of parenchymal cells by fibrous connective tissues. Fibroblast to myofibroblast transition (FMT) is considered to be the main source of myofibroblasts. Therefore, it is crucial to explore the influencing factors regulating the process of FMT for the prevention, treatment, and diagnosis of FMT-related diseases. In recent years, non-coding RNAs, including microRNA, long non-coding RNAs, and circular RNAs, have attracted extensive attention from scientists due to their powerful regulatory functions, and they have been found to play a vital role in regulating FMT. In this review, we summarized ncRNAs which regulate FMT during fibrosis and found that they mainly regulated signaling pathways, including TGF-β/Smad, MAPK/P38/ERK/JNK, PI3K/AKT, and WNT/β-catenin. Furthermore, the expression of downstream transcription factors can be promoted or inhibited, indicating that ncRNAs have the potential to be a new therapeutic target for FMT-related diseases.
Collapse
Affiliation(s)
| | | | | | - Jie Wang
- Department of Immunology, Xiangya School of Medicine, Central South University, Xiangya Road, Changsha 410000, China
| |
Collapse
|
5
|
Xia W, Li M, Jiang X, Huang X, Gu S, Ye J, Zhu L, Hou M, Zan T. Young fibroblast-derived exosomal microRNA-125b transfers beneficial effects on aged cutaneous wound healing. J Nanobiotechnology 2022; 20:144. [PMID: 35305652 PMCID: PMC9744129 DOI: 10.1186/s12951-022-01348-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Aged skin wounds heal poorly, resulting in medical, economic, and social burdens posed by nonhealing wounds. Age-related defects in repair are associated with reduced myofibroblasts and dysfunctional extracellular matrix (ECM) deposition. Bidirectional cell-cell communication involving exosome-borne cargo such as micro RNAs (miRs) has emerged as a critical mechanism for wound healing and aged tissue regeneration. Here we report that at the wound edge, aged fibroblasts display reduced migration and differentiation into myofibroblasts, with impaired ECM deposition, when compared with young tissue. Proper activation of fibroblasts to myofibroblasts may alleviate age-related defects in wound healing. Herein, an exosome-guided cell technique was performed to induce effective wound healing. Supplementing wounds with exosomes isolated from young mouse wound-edge fibroblasts (exosomesYoung) significantly improved the abundance of myofibroblasts and wound healing in aged mice and caused fibroblasts to migrate and transition to myofibroblasts in vitro. To determine the underlying mechanism, we found that exosomal transfer of miR-125b to fibroblasts inhibited sirtuin 7 (Sirt7), thus accelerating myofibroblast differentiation and wound healing in aged mice. Notably, after epidermal inhibition of miR-125b or overexpression of Sirt7 in fibroblasts, migration and myofibroblast transition were perturbed. Our findings thus reveal that miR-125b is transferred through exosomes from young fibroblasts to old fibroblasts contributes to promoting fibroblast migration and transition to counteract aging, suggesting a potential avenue for anti-aging interventions in wound healing.
Collapse
Affiliation(s)
- Wenzheng Xia
- grid.16821.3c0000 0004 0368 8293Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 200011 Shanghai, China
| | - Minxiong Li
- grid.16821.3c0000 0004 0368 8293Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 200011 Shanghai, China
| | - Xingyu Jiang
- grid.410745.30000 0004 1765 1045School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Huang
- grid.16821.3c0000 0004 0368 8293Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 200011 Shanghai, China
| | - Shuchen Gu
- grid.16821.3c0000 0004 0368 8293Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 200011 Shanghai, China
| | - Jiaqi Ye
- grid.268099.c0000 0001 0348 3990Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, No. 2 Fuxue Lane, 325000 Wenzhou, People’s Republic of China
| | - Liaoxiang Zhu
- grid.268099.c0000 0001 0348 3990Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, No. 2 Fuxue Lane, 325000 Wenzhou, People’s Republic of China
| | - Meng Hou
- grid.268099.c0000 0001 0348 3990Department of Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, No. 2 Fuxue Lane, 325000 Wenzhou, People’s Republic of China
| | - Tao Zan
- grid.16821.3c0000 0004 0368 8293Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 200011 Shanghai, China
| |
Collapse
|
6
|
Moscoso I, Cebro-Márquez M, Martínez-Gómez Á, Abou-Jokh C, Martínez-Monzonís MA, Martínez-Sande JL, González-Melchor L, García-Seara J, Fernández-López XA, Moraña-Fernández S, González-Juanatey JR, Rodríguez-Mañero M, Lage R. Circulating miR-499a and miR-125b as Potential Predictors of Left Ventricular Ejection Fraction Improvement after Cardiac Resynchronization Therapy. Cells 2022; 11:cells11020271. [PMID: 35053387 PMCID: PMC8773679 DOI: 10.3390/cells11020271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/24/2021] [Accepted: 01/06/2022] [Indexed: 11/30/2022] Open
Abstract
Cardiac resynchronization therapy represents a therapeutic option for heart failure drug-refractory patients. However, due to the lack of success in 30% of the cases, there is a demand for an in-depth analysis of individual heterogeneity. In this study, we aimed to evaluate the prognostic value of circulating miRNA differences. Responder patients were defined by a composite endpoint of the presence of left ventricular reverse remodelling (a reduction ≥15% in telesystolic volume and an increment ≥10% in left ventricular ejection fraction). Circulating miRNAs signature was analysed at the time of the procedure and at a 6-month follow-up. An expression analysis showed, both at baseline and at follow-up, differences between responders and non-responders. Responders presented lower baseline expressions of miR-499, and at follow-up, downregulation of miR-125b-5p, both associated with a significant improvement in left ventricular ejection fraction. The miRNA profile differences showed a marked sensitivity to distinguish between responders and non-responders. Our data suggest that miRNA differences might contribute to prognostic stratification of patients undergoing cardiac resynchronization therapy and suggest that preimplant cardiac context as well as remodelling response are key to therapeutic success.
Collapse
Affiliation(s)
- Isabel Moscoso
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (I.M.); (M.C.-M.); (J.R.G.-J.)
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - María Cebro-Márquez
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (I.M.); (M.C.-M.); (J.R.G.-J.)
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - Álvaro Martínez-Gómez
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - Charigan Abou-Jokh
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - María Amparo Martínez-Monzonís
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - José Luis Martínez-Sande
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Laila González-Melchor
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - Javier García-Seara
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Xesús Alberte Fernández-López
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - Sandra Moraña-Fernández
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
| | - José R. González-Juanatey
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (I.M.); (M.C.-M.); (J.R.G.-J.)
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Moisés Rodríguez-Mañero
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Ricardo Lage
- Cardiology Group, Centre for Research in Molecular Medicine and Chronic Diseases (CIMUS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (I.M.); (M.C.-M.); (J.R.G.-J.)
- Department of Cardiology and Coronary Unit and Cellular and Molecular Cardiology Research Unit, Institute of Biomedical Research (IDIS-SERGAS), University Clinical Hospital, 15706 Santiago de Compostela, Spain; (Á.M.-G.); (C.A.-J.); (M.A.M.-M.); (J.L.M.-S.); (L.G.-M.); (J.G.-S.); (X.A.F.-L.); (S.M.-F.); (M.R.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
7
|
Wang Y, Tan J, Wang L, Pei G, Cheng H, Zhang Q, Wang S, He C, Fu C, Wei Q. MiR-125 Family in Cardiovascular and Cerebrovascular Diseases. Front Cell Dev Biol 2021; 9:799049. [PMID: 34926475 PMCID: PMC8674784 DOI: 10.3389/fcell.2021.799049] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular and cerebrovascular diseases are a serious threaten to the health of modern people. Understanding the mechanism of occurrence and development of cardiovascular and cerebrovascular diseases, as well as reasonable prevention and treatment of them, is a huge challenge that we are currently facing. The miR-125 family consists of hsa-miR-125a, hsa-miR-125b-1 and hsa-miR-125b-2. It is a kind of miRNA family that is highly conserved among different species. A large amount of literature shows that the lack of miR-125 can cause abnormal development of the cardiovascular system in the embryonic period. At the same time, the miR-125 family participates in the occurrence and development of a variety of cardiovascular and cerebrovascular diseases, including myocardial ischemia, atherosclerosis, ischemia-reperfusion injury, ischemic stroke, and heart failure directly or indirectly. In this article, we summarized the role of the miR-125 family in the development and maturation of cardiovascular system, the occurrence and development of cardiovascular and cerebrovascular diseases, and its important value in the current fiery stem cell therapy. In addition, we presented this in the form of table and diagrams. We also discussed the difficulties and challenges faced by the miR-125 family in clinical applications.
Collapse
Affiliation(s)
- Yang Wang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Jing Tan
- Department of Ultrasound Medicine, Binzhou People's Hospital, Binzhou, China
| | - Lu Wang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Gaiqin Pei
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Hongxin Cheng
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Qing Zhang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Shiqi Wang
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Chenying Fu
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| |
Collapse
|
8
|
Jung HN, Jung CH. The Role of Anti-Inflammatory Adipokines in Cardiometabolic Disorders: Moving beyond Adiponectin. Int J Mol Sci 2021; 22:ijms222413529. [PMID: 34948320 PMCID: PMC8707770 DOI: 10.3390/ijms222413529] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023] Open
Abstract
The global burden of obesity has multiplied owing to its rapidly growing prevalence and obesity-related morbidity and mortality. In addition to the classic role of depositing extra energy, adipose tissue actively interferes with the metabolic balance by means of secreting bioactive compounds called adipokines. While most adipokines give rise to inflammatory conditions, the others with anti-inflammatory properties have been the novel focus of attention for the amelioration of cardiometabolic complications. This review compiles the current evidence on the roles of anti-inflammatory adipokines, namely, adiponectin, vaspin, the C1q/TNF-related protein (CTRP) family, secreted frizzled-related protein 5 (SFRP5), and omentin-1 on cardiometabolic health. Further investigations on the mechanism of action and prospective human trials may pave the way to their clinical application as innovative biomarkers and therapeutic targets for cardiovascular and metabolic disorders.
Collapse
Affiliation(s)
- Han Na Jung
- Asan Medical Center, Department of Internal Medicine, University of Ulsan College of Medicine, Seoul 05505, Korea;
- Asan Diabetes Center, Asan Medical Center, Seoul 05505, Korea
| | - Chang Hee Jung
- Asan Medical Center, Department of Internal Medicine, University of Ulsan College of Medicine, Seoul 05505, Korea;
- Asan Diabetes Center, Asan Medical Center, Seoul 05505, Korea
- Correspondence:
| |
Collapse
|
9
|
Huang X, Yan Y, Zheng W, Ma Y, Wang X, Gong W, Nie S. Secreted Frizzled-Related Protein 5 Protects Against Cardiac Rupture and Improves Cardiac Function Through Inhibiting Mitochondrial Dysfunction. Front Cardiovasc Med 2021; 8:682409. [PMID: 34568442 PMCID: PMC8458704 DOI: 10.3389/fcvm.2021.682409] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Secreted frizzled-related protein 5 (Sfrp5) has been suggested to be a protective regulatory protein in coronary heart disease. However, the role of Sfrp5 in regulating ischemic injury and its consequences is not known. The aim of our study was to explore the effects of Sfrp5 on hearts after myocardial infarction (MI) and to investigate the underlying mechanisms. Methods and Results: We found that Sfrp5 was downregulated over time in the heart tissue of MI mice. To further elucidate the role of Sfrp5 during MI, we established a cardiac overexpression of an Sfrp5 mouse model using the cardiotropic adeno-associated virus serotype 9 (AAV9). Overexpression of Sfrp5 significantly reduced infarct size as demonstrated by a decrease in mortality owing to cardiac rupture. Moreover, cardiac overexpression of Sfrp5 increased left ventricular function and mitochondrial biogenesis, decreased cardiomyocyte apoptosis, suppressed inflammation reaction, inhibited oxidative stress, and ameliorated cardiac remodeling as demonstrated by left ventricular ejection fraction, mitochondrial morphology, heart weight, NADH oxidase activity levels, and myocardial fibrosis at 2 weeks post-MI. At the molecular level, overexpression of Sfrp5 significantly increased the expression of p-AMPKThr172 protein with higher expression of mitochondrial fusion protein (MFN1 and MFN2) and lower expression of mitochondrial fission protein (p-Drp1Ser616/Mid49/MFF/Fis-1). In isolated neonatal rat cardiac myocytes, Sfrp5 treatment attenuated hypoxia-induced mitochondrial dysfunction. Inhibition of AMPK activity with compound C abrogated this benefit. Conclusions: Sfrp5 overexpression inhibits ischemic injury, reduces risk of cardiac rupture, ameliorates post-MI remodeling, and decreases the progression to heart failure via disrupting mitochondrial dysfunction and partly through normalizing the AMPK activity.
Collapse
Affiliation(s)
- Xin Huang
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Yan Yan
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Wen Zheng
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Youcai Ma
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Xiao Wang
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Wei Gong
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| | - Shaoping Nie
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.,Beijing Institute of Heart, Lung, and Blood Vessel Diseases, Beijing, China
| |
Collapse
|
10
|
MicroRNAs-The Heart of Post-Myocardial Infarction Remodeling. Diagnostics (Basel) 2021; 11:diagnostics11091675. [PMID: 34574016 PMCID: PMC8469128 DOI: 10.3390/diagnostics11091675] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 09/02/2021] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
Myocardial infarction (MI) is one of the most frequent cardiac emergencies, with significant potential for mortality. One of the major challenges of the post-MI healing response is that replacement fibrosis could lead to left ventricular remodeling (LVR) and heart failure (HF). This process involves canonical and non-canonical transforming growth factor-beta (TGF-β) signaling pathways translating into an intricate activation of cardiac fibroblasts and disproportionate collagen synthesis. Accumulating evidence has indicated that microRNAs (miRNAs) significantly contribute to the modulation of these signaling pathways. This review summarizes the recent updates regarding the molecular mechanisms underlying the role of the over 30 miRNAs involved in post-MI LVR. In addition, we compare the contradictory roles of several multifunctional miRNAs and highlight their potential use in pressure overload and ischemia-induced fibrosis. Finally, we discuss their attractive role as prognostic biomarkers for HF, highlighting the most relevant human trials involving these miRNAs.
Collapse
|
11
|
Zhang B, Mao S, Liu X, Li S, Zhou H, Gu Y, Liu W, Fu L, Liao C, Wang P. MiR-125b inhibits cardiomyocyte apoptosis by targeting BAK1 in heart failure. Mol Med 2021; 27:72. [PMID: 34238204 PMCID: PMC8268255 DOI: 10.1186/s10020-021-00328-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 06/10/2021] [Indexed: 01/27/2023] Open
Abstract
Background Although miR-125b plays a crucial role in many human cancers. However, its function in heart failure (HF) remains unclear. Our study aimed to investigate its involvement in heart failure. Methods In this study, the mouse HF model was successfully constructed through transverse aortic constriction (TAC) operation. Changes in mRNA and protein levels in isolated myocytes and heart tissues were examined using qRT-PCR, Western blot and Immunohistochemical staining and immunofluorescent staining. Changes in cardiac functions were examined using ultrasound. Interactions between miR-125b and BAK1 was analyzed using the luciferase reporter assay. Cardiomyocyte apoptosis was evaluated using the TUNEL staining. Results We found that miR-125b expression was significantly downregulated in myocardial tissues of HF mice. Moreover, miR-125b upregulation in HF mice injected with agomir-125b efficiently ameliorated cardiac function. Further, miR-125b upregulation significantly decreased the protein levels of apoptosis-related makers c-caspase 3 and Bax, while increased Bcl-2 expression. In addition, BAK1 was identified as a direct target of miR-125b. As expected, BAK1 overexpression observably reversed the effect of agomir-125b on cardiac function and on the expression of apoptosis-related makers in the heart tissues of HF mice. Conclusions Taken together, miR-125b overexpression efficiently attenuated cardiac function injury of HF mice by targeting BAK1 through inhibiting cardiomyocyte apoptosis, suggesting that miR-125b/BAK1 axis might be a potential target for the diagnosis or treatment of HF. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-021-00328-w.
Collapse
Affiliation(s)
- Bei Zhang
- Guizhou Medical University, No. 9 Beijing Road, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China.,Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China
| | - Shanyong Mao
- Guizhou Medical University, No. 9 Beijing Road, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China.,Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China
| | - Xingde Liu
- Guizhou Medical University, No. 9 Beijing Road, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China. .,Department of Cardiology, The Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, 550004, People's Republic of China.
| | - Sha Li
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China.
| | - Haiyan Zhou
- Departmentof Clinical Research Centre, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Ying Gu
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China
| | - Wupeng Liu
- Department of Cardiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guiyang City, Guizhou, 550014, People's Republic of China
| | - Lei Fu
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China
| | - Chunyan Liao
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China
| | - Pengzhen Wang
- Department of Ultrasound Medicine, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Street, Yunyan District, Guiyang, Guizhou, 550004, People's Republic of China
| |
Collapse
|
12
|
Yang T, Long T, Du T, Chen Y, Dong Y, Huang ZP. Circle the Cardiac Remodeling With circRNAs. Front Cardiovasc Med 2021; 8:702586. [PMID: 34250050 PMCID: PMC8267062 DOI: 10.3389/fcvm.2021.702586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/02/2021] [Indexed: 12/16/2022] Open
Abstract
Cardiac remodeling occurs after the heart is exposed to stress, which is manifested by pathological processes such as cardiomyocyte hypertrophy and apoptosis, dendritic cells activation and cytokine secretion, proliferation and activation of fibroblasts, and finally leads to heart failure. Circular RNAs (circRNAs) are recently recognized as a specific type of non-coding RNAs that are expressed in different species, in different stages of development, and in different pathological conditions. Growing evidences have implicated that circRNAs play important regulatory roles in the pathogenesis of a variety of cardiovascular diseases. In this review, we summarize the biological origin, characteristics, functional classification of circRNAs and their regulatory functions in cardiomyocytes, endothelial cells, fibroblasts, immune cells, and exosomes in the pathogenesis of cardiac remodeling.
Collapse
Affiliation(s)
- Tiqun Yang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Tianxin Long
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Tailai Du
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yili Chen
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission (NHC) Key Laboratory of Assisted Circulation, Sun Yat-sen University, Guangzhou, China.,National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, China
| |
Collapse
|
13
|
Lin F, Zhang S, Liu X, Wu M. RETRACTED: Mouse bone marrow derived mesenchymal stem cells-secreted exosomal microRNA-125b-5p suppresses atherosclerotic plaque formation via inhibiting Map4k4. Life Sci 2021; 274:119249. [PMID: 33652034 DOI: 10.1016/j.lfs.2021.119249] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 01/16/2021] [Accepted: 01/25/2021] [Indexed: 02/08/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor-in-Chief. Concern was raised about the reliability of the Western blot results in Figs. 2D and 4E, which appear to have the same eyebrow shaped phenotype as many other publications tabulated here (https://docs.google.com/spreadsheets/d/149EjFXVxpwkBXYJOnOHb6RhAqT4a2llhj9LM60MBffM/edit#gid=0). The journal requested the corresponding author comment on these concerns and provide the raw data. However the authors were not able to satisfactorily fulfil this request and therefore the Editor-in-Chief decided to retract the article.
Collapse
Affiliation(s)
- Feng Lin
- Department of Cardiology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518000, Guangdong, China.
| | - Suihao Zhang
- Department of Cardiology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518000, Guangdong, China
| | - Xia Liu
- Department of Cardiology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518000, Guangdong, China
| | - Meishan Wu
- Department of Cardiology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518000, Guangdong, China
| |
Collapse
|
14
|
Zhu M, Liu X, Li W, Wang L. Exosomes derived from mmu_circ_0000623-modified ADSCs prevent liver fibrosis via activating autophagy. Hum Exp Toxicol 2020; 39:1619-1627. [PMID: 32633558 DOI: 10.1177/0960327120931152] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Prolonged parenchymal cell death leads to activation of fibrogenic cells, extracellular matrix accumulation, and eventually liver fibrosis. Increasing evidence shows that exosomes (Exos) secreted by adipose-derived mesenchymal stem cells (ADSCs) can be used to deliver circular RNAs (circRNAs) to treat liver fibrosis. To explore the uses of circRNA, circRNA expression profiles of hepatic tissue from normal and CCl4-induced mice were analyzed using high-throughput circRNA microarrays. The result showed that mmu_circ_0000623 expression was downregulated in CCl4-induced mice. Bioinformatics analysis and luciferin reporter experiments showed that mmu_circ_0000623 interacted with and regulated miR-125/ATG4D. In vitro and in vivo experiments showed that Exos from ADSCs, especially from mmu_circ_0000623-modified ADSCs, significantly suppressed CCl4-induced liver fibrosis by promoting autophagy activation. Autophagy inhibitor treatment significantly reversed the treatment effects of Exos. Proteins involved in autophagy and autophagy plaques positive for ATG4D expression were regulated by mmu_circ_0000623/miR-125. Our study found that Exos derived from mmu_circ_0000623-modified ADSCs prevented liver fibrosis via activating autophagy.
Collapse
Affiliation(s)
- M Zhu
- Geriatric Department, 117947Jining No. 1 People's Hospital, Jining, Shandong, China
| | - X Liu
- Geriatric Department, 117947Jining No. 1 People's Hospital, Jining, Shandong, China
| | - W Li
- Geriatric Department, 117947Jining No. 1 People's Hospital, Jining, Shandong, China
| | - L Wang
- Geriatric Department, 117947Jining No. 1 People's Hospital, Jining, Shandong, China
| |
Collapse
|
15
|
Wang B, Pan Y, Yang G, Cui Z, Yu W, Liu H, Bai B. Sfrp5/Wnt5a and leptin/adiponectin levels in the serum and the periarterial adipose tissue of patients with peripheral arterial occlusive disease. Clin Biochem 2020; 87:46-51. [PMID: 33188773 DOI: 10.1016/j.clinbiochem.2020.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 10/15/2020] [Accepted: 11/03/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Leptin, adiponectin, secreted frizzled-related protein 5 (Sfrp5) and wingless-type family member 5a (Wnt5a) are novel adipokines that are involved in insulin sensitivity and atherosclerosis. The aim of the present study was to investigate the serum and periarterial adipose tissue leptin/adiponectin and Sfrp5/Wnt5a levels in patients with peripheral arterial occlusive disease (PAOD). METHODS A total of 75 patients with PAOD and 39 control subjects were recruited. The serum concentrations of leptin, adiponectin, Sfrp5 and Wnt5a were measured by ELISAs, and the leptin, adiponectin, Sfrp5 and Wnt5a levels in the periarterial adipose tissue were observed by western blotting. RESULTS The serum Sfrp5 levels were significantly lower in the patients with PAOD than in the control subjects (p < 0.001) and Wnt5a levels were higher in the patients with PAOD (p < 0.001). The serum leptin levels were significantly higher in the patients with PAOD than in the control subjects (p < 0.001), and adiponectin levels were significantly lower in the patients with PAOD (p < 0.001). The serum Sfrp5 levels were associated with ABI (rs = 0.274; p = 0.018), Wnt5a (rs = -0.409; p < 0.001), adiponectin (rs = 0.244; p = 0.035) and Leptin/Adiponetin ratio (rs = -0.244; p = 0.037). The adiponectin and Sfrp5 protein levels were decreased in the periarterial adipose tissue of patients with PAOD compared with control subjects. The leptin and Wnt5a protein levels were increased in the periarterial adipose tissue of patients with PAOD compared with control subjects. CONCLUSION We demonstrated that the adiponectin and Sfrp5 levels in the serum and periarterial adipose tissue were significantly lower in the patients with PAOD than in the control subjects. The leptin and Wnt5a levels in the serum and periarterial adipose tissue were significantly higher in the patients with PAOD than in the control subjects.
Collapse
Affiliation(s)
- Biyu Wang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu Pan
- The Department of Medical Administration, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guang Yang
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zong'ao Cui
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenping Yu
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Liu
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bing Bai
- The Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
16
|
Wu J, Zheng H, Liu X, Chen P, Zhang Y, Luo J, Kuang J, Li J, Yang Y, Ma T, Yang Y, Huang X, Liang G, Liang D, Hu Y, Wu JHY, Arnott C, Mai W, Huang Y. Prognostic Value of Secreted Frizzled-Related Protein 5 in Heart Failure Patients With and Without Type 2 Diabetes Mellitus. Circ Heart Fail 2020; 13:e007054. [PMID: 32842761 DOI: 10.1161/circheartfailure.120.007054] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/12/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Patients with heart failure (HF) with diabetes mellitus have distinct biomarker profiles compared with those without diabetes mellitus. SFRP5 (secreted frizzled-related protein 5) is an anti-inflammatory adipokine with an important suppressing role on the development of type 2 diabetes mellitus (T2DM). This study aimed to evaluate the prognostic value of SFRP5 in patients with HF with and without T2DM. METHODS The study included 833 consecutive patients with HF, 312 (37.5%) of whom had T2DM. Blood samples were collected at presentation, and SFRP5 levels were measured. The primary outcome was the composite end points of first occurrence of HF rehospitalization or all-cause mortality during follow-up. RESULTS During median follow-up of 2.1 years, 335 (40.2%) patients in the cohort experienced the composite primary outcome. After adjustment for multiple risk factors, each doubling of SFRP5 level was associated with a 21% decreased risk of primary outcomes in the overall study population (P<0.001). Subgroup analyses showed that the association between level of SFPR5 and primary outcomes may be stronger in patients with T2DM (hazard ratio, 0.69 [95% CI, 0.61-0.79]) than in patients without T2DM (hazard ratio, 0.89 [95% CI, 0.79-1.01]; interaction P=0.006). Similar associations were observed when taking SFRP5 as a categorical variable. Addition of SFRP5 significantly improved discrimination and reclassification of the incident primary outcomes beyond clinical risk factors and N-terminal pro-B-type natriuretic peptide in all patients with HF and those with T2DM (all P<0.01). CONCLUSIONS SFRP5 is an independent novel biomarker for risk stratification in HF, especially in HF with T2DM.
Collapse
Affiliation(s)
- Jiandi Wu
- Department of Cardiology, Affiliated Foshan Hospital (J.W., D.L.), Southern Medical University, Foshan, China
| | - Haoxiao Zheng
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
| | - Xinyue Liu
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
| | - Peisong Chen
- Department of Laboratory medicine (P.C.), the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yunlong Zhang
- Key Laboratory of Neuroscience, School of Basic Medical Sciences, Guangzhou Medical University, China (Y.Z.)
| | - Jianjin Luo
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
- Department of Cardiology, the Second Hospital of Zhaoqing, Guangdong, China (J. Luo, G.L.)
| | - Jian Kuang
- Department of Cardiology (J.K., W.M.), the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), China (J.K., W.M.)
| | - Jingwei Li
- Department of Cardiology, People's Liberation Army General Hospital, Beijing, China (J. Li)
| | - Yu Yang
- Department of Geriatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China (Yu Yang)
| | - Tianyi Ma
- Department of Cardiology, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Changsha, China (T.M.)
| | - Yanhua Yang
- Department of Cardiology, Dongguan people's Hospital (Yanhua Yang), Southern Medical University, Foshan, China
| | - Xiaohui Huang
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
| | - Guoquan Liang
- Department of Cardiology, the Second Hospital of Zhaoqing, Guangdong, China (J. Luo, G.L.)
| | - Donglian Liang
- Department of Cardiology, Affiliated Foshan Hospital (J.W., D.L.), Southern Medical University, Foshan, China
| | - Yunzhao Hu
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
| | - Jason H Y Wu
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (J.H.Y.W., C.A., Y. Huang)
| | - Clare Arnott
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (J.H.Y.W., C.A., Y. Huang)
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, Australia (C.A.)
- Charles Perkins Centre, University of Sydney, NSW, Australia (C.A.)
| | - Weiyi Mai
- Department of Cardiology (J.K., W.M.), the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), China (J.K., W.M.)
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital (H.Z., X.L., J. Luo, X.H., Y. Hu, Y. Huang), Southern Medical University, Foshan, China
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, Australia (J.H.Y.W., C.A., Y. Huang)
| |
Collapse
|
17
|
Hu HH, Cao G, Wu XQ, Vaziri ND, Zhao YY. Wnt signaling pathway in aging-related tissue fibrosis and therapies. Ageing Res Rev 2020; 60:101063. [PMID: 32272170 DOI: 10.1016/j.arr.2020.101063] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/25/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Fibrosis is the final hallmark of pathological remodeling, which is a major contributor to the pathogenesis of various chronic diseases and aging-related organ failure to fully control chronic wound-healing and restoring tissue function. The process of fibrosis is involved in the pathogenesis of the kidney, lung, liver, heart and other tissue disorders. Wnt is a highly conserved signaling in the aberrant wound repair and fibrogenesis, and sustained Wnt activation is correlated with the pathogenesis of fibrosis. In particular, mounting evidence has revealed that Wnt signaling played important roles in cell fate determination, proliferation and cell polarity establishment. The expression and distribution of Wnt signaling in different tissues vary with age, and these changes have key effects on maintaining tissue homeostasis. In this review, we first describe the major constituents of the Wnt signaling and their regulation functions. Subsequently, we summarize the dysregulation of Wnt signaling in aging-related fibrotic tissues such as kidney, liver, lung and cardiac fibrosis, followed by a detailed discussion of its involvement in organ fibrosis. In addition, the crosstalk between Wnt signaling and other pathways has the potential to profoundly add to the complexity of organ fibrosis. Increasing studies have demonstrated that a number of Wnt inhibitors had the potential role against tissue fibrosis, specifically in kidney fibrosis and the implications of Wnt signaling in aging-related diseases. Therefore, targeting Wnt signaling might be a novel and promising therapeutic strategy against aging-related tissue fibrosis.
Collapse
Affiliation(s)
- He-He Hu
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Xia-Qing Wu
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, Irvine, California, 92897, USA
| | - Ying-Yong Zhao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
18
|
Szabó MR, Gáspár R, Pipicz M, Zsindely N, Diószegi P, Sárközy M, Bodai L, Csont T. Hypercholesterolemia Interferes with Induction of miR-125b-1-3p in Preconditioned Hearts. Int J Mol Sci 2020; 21:ijms21113744. [PMID: 32466450 PMCID: PMC7312064 DOI: 10.3390/ijms21113744] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/19/2022] Open
Abstract
Ischemic preconditioning (IPre) reduces ischemia/reperfusion (I/R) injury in the heart. The non-coding microRNA miR-125b-1-3p has been demonstrated to play a role in the mechanism of IPre. Hypercholesterolemia is known to attenuate the cardioprotective effect of preconditioning; nevertheless, the exact underlying mechanisms are not clear. Here we investigated, whether hypercholesterolemia influences the induction of miR-125b-1-3p by IPre. Male Wistar rats were fed with a rodent chow supplemented with 2% cholesterol and 0.25% sodium-cholate hydrate for 8 weeks to induce high blood cholesterol levels. The hearts of normo- and hypercholesterolemic animals were then isolated and perfused according to Langendorff, and were subjected to 35 min global ischemia and 120 min reperfusion with or without IPre (3 × 5 min I/R cycles applied before index ischemia). IPre significantly reduced infarct size in the hearts of normocholesterolemic rats; however, IPre was ineffective in the hearts of hypercholesterolemic animals. Similarly, miR-125b-1-3p was upregulated by IPre in hearts of normocholesterolemic rats, while in the hearts of hypercholesterolemic animals IPre failed to increase miR-125b-1-3p significantly. Phosphorylation of cardiac Akt, ERK, and STAT3 was not significantly different in any of the groups at the end of reperfusion. Based on these results we propose here that hypercholesterolemia attenuates the upregulation of miR-125b-1-3p by IPre, which seems to be associated with the loss of cardioprotection.
Collapse
Affiliation(s)
- Márton R. Szabó
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9., H-6720 Szeged, Hungary; (M.R.S.); (R.G.); (M.P.); (P.D.); (M.S.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13., H-6720 Szeged, Hungary
| | - Renáta Gáspár
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9., H-6720 Szeged, Hungary; (M.R.S.); (R.G.); (M.P.); (P.D.); (M.S.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13., H-6720 Szeged, Hungary
| | - Márton Pipicz
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9., H-6720 Szeged, Hungary; (M.R.S.); (R.G.); (M.P.); (P.D.); (M.S.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13., H-6720 Szeged, Hungary
| | - Nóra Zsindely
- Department of Microbiology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52., H-6726 Szeged, Hungary;
| | - Petra Diószegi
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9., H-6720 Szeged, Hungary; (M.R.S.); (R.G.); (M.P.); (P.D.); (M.S.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13., H-6720 Szeged, Hungary
| | - Márta Sárközy
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9., H-6720 Szeged, Hungary; (M.R.S.); (R.G.); (M.P.); (P.D.); (M.S.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13., H-6720 Szeged, Hungary
| | - László Bodai
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Közép fasor 52., H-6726 Szeged, Hungary;
| | - Tamás Csont
- Metabolic Diseases and Cell Signaling (MEDICS) Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Dóm tér 9., H-6720 Szeged, Hungary; (M.R.S.); (R.G.); (M.P.); (P.D.); (M.S.)
- Interdisciplinary Centre of Excellence, University of Szeged, Dugonics tér 13., H-6720 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-096
| |
Collapse
|
19
|
Tong S, Du Y, Ji Q, Dong R, Cao J, Wang Z, Li W, Zeng M, Chen H, Zhu X, Zhou Y. Expression of Sfrp5/Wnt5a in human epicardial adipose tissue and their relationship with coronary artery disease. Life Sci 2020; 245:117338. [PMID: 31981630 DOI: 10.1016/j.lfs.2020.117338] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 01/03/2020] [Accepted: 01/18/2020] [Indexed: 12/11/2022]
Abstract
Secreted frizzled-related protein 5 (Sfrp5) primarily acts in combination with wingless-type family member 5a (Wnt5a), to inhibits chronic inflammation and repress atherosclerosis and other metabolic disorders. Epicardial adipose tissue (EAT), surrounding the heart and coronary arteries, has been found to be highly related to the progression of coronary artery disease through adipokines production. However, little is known about EAT-derived Sfrp5 and Wnt5a in humans. We aimed to investigate whether the EAT-derived Sfrp5/Wnt5a levels are altered in patients with CAD. Fifty-eight patients with CAD and 29 patients without CAD who underwent cardiac surgery were enrolled. Serum samples and paired adipose biopsies from EAT and subcutaneous adipose tissue (SAT) were collected, and Sfrp5 and Wnt5a levels were detected. Correlation and multivariate regression analyses were performed to determine the relationship between Sfrp5/Wnt5a expression and CAD and other clinical risk factors. According to the results, the CAD group had lower Sfrp5 and higher Wnt5a levels in EAT and serum (all p < 0.05). Serum Sfrp5 levels were significantly lower in CAD patients with impaired myocardial function. EAT Sfrp5 mRNA levels and serum Sfrp5 levels were both negatively associated with the presence of CAD, after adjustment for known biomarkers, EAT mRNA and serum Wnt5a levels correlated positively with the presence of CAD. Thus, we concluded that low Sfrp5 and high Wnt5a levels are associated with the presence of CAD, independent of other conventional risk factors.
Collapse
Affiliation(s)
- Shan Tong
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China; Center of Geriatrics, Hainan General Hospital, Hainan 580000, China
| | - Yu Du
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China
| | - Qingwei Ji
- Emergency & Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ran Dong
- Department of Cardiac Surgery Center, 11th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Jian Cao
- Department of Cardiac Surgery Center, 11th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Zhijian Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China
| | - Wei Li
- Center of Geriatrics, Hainan General Hospital, Hainan 580000, China
| | - Min Zeng
- Center of Geriatrics, Hainan General Hospital, Hainan 580000, China
| | - Hongying Chen
- The Jackson Clinics, Physical Therapy, Middleburg, VA 20117, USA
| | - Xiaogang Zhu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China
| | - Yujie Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing 100029, China.
| |
Collapse
|
20
|
Sun L, Zhao J, Ge X, Zhang H, Wang C, Bie Z. Circ_LAS1L regulates cardiac fibroblast activation, growth, and migration through miR‐125b/SFRP5 pathway. Cell Biochem Funct 2020; 38:443-450. [PMID: 31950540 DOI: 10.1002/cbf.3486] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Li‐ye Sun
- Department of GeratologyYantai Yuhuangding Hospital Yantai China
| | - Jin‐chao Zhao
- Department of Peripheral Vascular SurgeryDongzhimen Hospital, Beijing University of Chinese Medicine Beijing China
| | - Xiao‐ming Ge
- Department of CardiologyThe First Hospital of Fangshan District Beijing China
| | - Hui Zhang
- Department of CardiologyFeixian People's Hospital, Shandong Medical College Linyi China
| | - Chuan‐mei Wang
- Department of CardiologyFeixian People's Hospital, Shandong Medical College Linyi China
| | - Zi‐dong Bie
- Department of CardiologyFeixian People's Hospital, Shandong Medical College Linyi China
- Department of CardiologyMeishanCardio‐Cerebrovascular Disease Hospital Meishan China
| |
Collapse
|
21
|
Du Y, Zhao Y, Zhu Y, Hu C, Zhang J, Ji Q, Liu W, Han H, Yang L, Zhang D, Tong S, Wang Z, Guo Y, Liu X, Zhou Y. High Serum Secreted Frizzled-Related Protein 5 Levels Associates with Early Improvement of Cardiac Function Following ST-Segment Elevation Myocardial Infarction Treated by Primary Percutaneous Coronary Intervention. J Atheroscler Thromb 2019; 26:868-878. [PMID: 30773518 PMCID: PMC6800391 DOI: 10.5551/jat.47019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023] Open
Abstract
AIM Several members of secreted frizzled-related protein (SFRP) are involved in the process of myocardial ischemia-reperfusion injury. However, little is known about the role of SFRP5 in patients with acute ST-segment elevation myocardial infarction (STEMI). METHODS In this cross-sectional study, 85 patients with first-time anterior STEMI who underwent timely primary percutaneous coronary intervention (PCI) and 35 patients without coronary artery disease (CAD) were enrolled. Serum SFRP5 levels were measured using an enzyme-linked immunosorbent assay kit. Patients with STEMI were divided into low-SFRP5 and high-SFRP5 groups according to their median baseline serum SFRP5 levels. To evaluate cardiac function and structure after infarction, the left ventricular ejection fraction (LVEF) and left ventricular end-diastolic volume (LVEDV) were measured using echocardiography. The associations between changes in LVEF and reduced LVEF (≤ 50%) and clinical variables were determined by univariate and multivariate analyses. RESULTS Baseline serum SFRP5 levels were significantly higher in patients with STEMI than in those without CAD (23.3 ng/mL vs 19.8 ng/mL, P=0.008), although they decreased over time. Also, baseline serum SFRP5 levels were inversely correlated with peak hypersensitive cardiac troponin I (hs-cTnI) levels (r=-0.234, P=0.025) and peak hypersensitive C-reactive protein (hs-CRP) levels (r=-0.262, P=0.015). A multivariate linear regression model showed that changes in LVEF were positively correlated with serum SFRP5 levels at baseline (β= 0.249, 95% confidence interval (CI) 0.018-0.245, P=0.024) and 24 h after admission (β=0.220, 95% CI 0.003-0.264, P=0.045). At 3 months, LVEF in patients with high SFRP5 levels was significantly improved over baseline [(60.8±7.1) % vs (56.1±7.5) %, P=0.001]. LVEF was also significantly higher in patients with high SFRP5 levels than in those with low at the 3-month follow-up [(60.8±7.1) % vs (56.8±8.9) %, P=0.028]. Consequently, high serum SFRP5 levels at baseline were associated with a decreased risk of reduced LVEF at 3 months, independent of peak hs-cTnI and baseline cardiac function (hazard ratio 0.190, 95% CI 0.036-0.996; P=0.049). CONCLUSIONS High serum SFRP5 levels measured during the acute phase of STEMI were significantly associated with promoting myocardial recovery at an early phase following primary PCI, suggesting that SFRP5 is a potential therapeutic target in acute STEMI.
Collapse
Affiliation(s)
- Yu Du
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yingxin Zhao
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yong Zhu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Chenping Hu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Jianwei Zhang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Qingwei Ji
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Wei Liu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Hongya Han
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Lixia Yang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Dai Zhang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Shan Tong
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Zhijian Wang
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yonghe Guo
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Xiaoli Liu
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| | - Yujie Zhou
- Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Clinical Center for Coronary Heart Disease, Beijing, China
| |
Collapse
|
22
|
Ghasemi A, Hashemy SI, Azimi-Nezhad M, Dehghani A, Saeidi J, Mohtashami M. The cross-talk between adipokines and miRNAs in health and obesity-mediated diseases. Clin Chim Acta 2019; 499:41-53. [PMID: 31476303 DOI: 10.1016/j.cca.2019.08.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 08/28/2019] [Accepted: 08/28/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Multiple studies have revealed a direct correlation between obesity and the development of multiple comorbidities, including metabolic diseases, cardiovascular disorders, chronic inflammatory disease, and cancers. However, the molecular mechanism underlying the link between obesity and the progression of these diseases is not completely understood. Adipokines are factors that are secreted by adipocytes and play a key role in whole body homeostasis. Collaboratively, miRNAs are suggested to have key functions in the development of obesity and obesity-related disorders. Based on recently emerging evidence, obesity leads to the dysregulation of both adipokines and obesity-related miRNAs. In the present study, we described the correlations between obesity and its related diseases that are mediated by the mutual regulatory effects of adipokines and miRNAs. METHODS We reviewed current knowledge of the modulatory effects of adipokines on miRNAs activity and their relevant functions in pathological conditions and vice versa. RESULTS Our research reveals the ability of adipokines and miRNAs to control the expression and activity of the other class of molecules, and their effects on obesity-related diseases. CONCLUSIONS This study may help researchers develop a roadmap for future investigations and provide opportunities to develop new therapeutic and diagnostic methods for treating obesity-related diseases.
Collapse
Affiliation(s)
- Ahmad Ghasemi
- Non-communicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| | - Seyed Isaac Hashemy
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mohsen Azimi-Nezhad
- Non-communicable Disease Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran; UMR INSERM U 1122, IGE-PCV, Interactions Gène-Environment en Physiopathologie Cardiovascular Université de Lorraine, France
| | - Alireza Dehghani
- Institute of Biochemistry and Molecular Biology, University of Bonn, Bonn, Germany
| | - Jafar Saeidi
- Department of Physiology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Mahnaz Mohtashami
- Department of Biology, School of Basic Science, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| |
Collapse
|
23
|
Tong S, Ji Q, Du Y, Zhu X, Zhu C, Zhou Y. Sfrp5/Wnt Pathway: A Protective Regulatory System in Atherosclerotic Cardiovascular Disease. J Interferon Cytokine Res 2019; 39:472-482. [PMID: 31199714 PMCID: PMC6660834 DOI: 10.1089/jir.2018.0154] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 03/21/2019] [Indexed: 12/19/2022] Open
Abstract
Adipose tissue stores energy and is the largest endocrine organ in the body, producing several adipokines. However, among these adipokines, few play a role in the positive metabolism that promotes good health. Secreted frizzled-related protein (Sfrp)-5, an antagonist that directly binds to Wnt, has attracted interest due to its favorable effects on atherosclerotic cardiovascular disease (ASCVD). This review focuses on Sfrp5 biology and the roles of the Sfrp5/Wnt system in ASCVD.
Collapse
Affiliation(s)
- Shan Tong
- Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
- Department of Geriatric Medicine and Gerontology, Hainan General Hospital, Hainan, China
| | - Qingwei Ji
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yu Du
- Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Xiaogang Zhu
- Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| | - Caizhong Zhu
- Department of Geriatric Medicine and Gerontology, Hainan General Hospital, Hainan, China
| | - Yujie Zhou
- Beijing Key Laboratory of Precision Medicine of Coronary Atherosclerotic Disease, Department of Cardiology, 12th ward, Beijing Anzhen Hospital, Beijing Institute of Heart Lung and Blood Vessel Disease, Clinical Center for Coronary Heart Disease, Capital Medical University, Beijing, China
| |
Collapse
|
24
|
Abstract
Myocardial infarction and post-infarction left ventricular remodelling involve a high risk of morbidity and mortality. For this reason, ongoing research is being conducted in order to learn the mechanisms of unfavourable left ventricular remodelling following a myocardial infarction. New biomarkers are also being sought that would allow for early identification of patients with a high risk of post-infarction remodelling and dysfunction of the left ventricle. In recent years, there has been ever more experimental data that confirms the significance of microRNA in cardiovascular diseases. It has been confirmed that microRNAs are stable in systemic circulation, and can be directly measured in patients' blood. It has been found that significant changes occur in the concentrations of various types of microRNA in myocardial infarction and heart failure patients. Various types of microRNA are also currently being intensively researched in terms of their usefulness as markers of cardiomyocyte necrosis, and predictors of the post-infarction heart failure development. This paper is a summary of the current knowledge on the significance of microRNA in post-infarction left ventricular remodelling and heart failure.
Collapse
Affiliation(s)
- Mieczysław Dutka
- Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Faculty of Health Sciences, Willowa St. 2, 43-309, Bielsko-Biała, Poland.
| | - Rafał Bobiński
- Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Faculty of Health Sciences, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| | - Jan Korbecki
- Department of Biochemistry and Molecular Biology, University of Bielsko-Biala, Faculty of Health Sciences, Willowa St. 2, 43-309, Bielsko-Biała, Poland
| |
Collapse
|
25
|
Vu LT, Peng B, Zhang DX, Ma V, Mathey-Andrews CA, Lam CK, Kiomourtzis T, Jin J, McReynolds L, Huang L, Grimson A, Cho WC, Lieberman J, Le MT. Tumor-secreted extracellular vesicles promote the activation of cancer-associated fibroblasts via the transfer of microRNA-125b. J Extracell Vesicles 2019; 8:1599680. [PMID: 31044053 PMCID: PMC6484490 DOI: 10.1080/20013078.2019.1599680] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 02/15/2019] [Accepted: 03/21/2019] [Indexed: 12/12/2022] Open
Abstract
Tumour cells release large quantities of extracellular vesicles (EVs) to mediate their interactions with other cells in the tumour microenvironment. To identify host cells that naturally take up EVs from tumour cells, we created breast cancer cell lines secreting fluorescent EVs. These fluorescent EVs are taken up most robustly by fibroblasts within the tumour microenvironment. RNA sequencing indicated that miR-125b is one of the most abundant microRNAs secreted by mouse triple-negative breast cancer 4T1 and 4TO7 cells. Treatment with 4T1 EVs leads to an increase in fibroblast activation in isogenic 4TO7 tumours, which is reversed by blocking miR-125b in 4T1 EVs; hence, miR-125b delivery by EVs is responsible for fibroblast activation in mouse tumour models. miR-125b is also secreted by human breast cancer cells and the uptake of EVs from these cells significantly increases cellular levels of miR-125b and expression of multiple cancer-associated fibroblast markers in resident fibroblasts. Overexpression of miR-125b in both mouse and human fibroblasts leads to an activated phenotype similar to the knockdown of established miR-125b target mRNAs. These data indicate that miR-125b is transferred through EVs from breast cancer cells to normal fibroblasts within the tumour microenvironment and contributes to their development into cancer-associated fibroblasts.
Collapse
Affiliation(s)
- Luyen Tien Vu
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Boya Peng
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Daniel Xin Zhang
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | - Victor Ma
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Camille A. Mathey-Andrews
- Program in Cellular and Molecular Medicine, Boston Children‘s Hospital, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Chun Kuen Lam
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
| | | | | | | | - Linfeng Huang
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, P. R. China
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| | - Judy Lieberman
- Program in Cellular and Molecular Medicine, Boston Children‘s Hospital, and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Minh Tn Le
- Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, P. R. China
| |
Collapse
|
26
|
Wang XL, Zhao YY, Sun L, Shi Y, Li ZQ, Zhao XD, Xu CG, Ji HG, Wang M, Xu WR, Zhu W. Exosomes derived from human umbilical cord mesenchymal stem cells improve myocardial repair via upregulation of Smad7. Int J Mol Med 2018; 41:3063-3072. [PMID: 29484378 DOI: 10.3892/ijmm.2018.3496] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 02/09/2018] [Indexed: 12/13/2022] Open
Abstract
It has been previously reported that exosomes derived from human umbilical cord mesenchymal stem cells (hucMSC)‑exosomes exhibit cardioprotective effects on the rat acute myocardial infarction (AMI) models and cardiomyocyte hypoxia injury models in vitro, however the exact mechanisms involved require further investigation. The present study aimed to investigate the repair effects of hucMSC‑exosomes on myocardial injury via the regulation of mothers against decapentaplegic homolog 7 (Smad7) expression. Compared with sham or normoxia groups (in vivo and in vitro, respectively), western blotting demonstrated that Smad7 expression was significantly decreased in the borderline area of infraction myocardium and in H9C2(2‑1) cells following hypoxia‑induced injury. Additionally, microRNA (miR)‑125b‑5p expression was markedly increased using reverse transcription‑quantitative polymerase chain reaction, but was reversed by hucMSC‑exosomes. Trypan blue staining and lactate dehydrogenase release detection demonstrated that cell injury was significantly increased in the AMI + PBS and hypoxia group compared with in the sham and normoxia groups and was inhibited by hucMSC‑exosomes. A dual luciferase reporter gene assay confirmed that Smad7 is a target gene of miR‑125b‑5p. In addition, miR‑125b‑5p mimics promoted H9C2(2‑1) cell injury following 48 h exposure to hypoxia. Downregulation of Smad7 expression under hypoxia was increased by miR‑125b‑5p mimics compared with the mimic negative control, and hucMSC‑exosomes partially alleviated this phenomenon. In conclusion, hucMSC‑exosomes may promote Smad7 expression by inhibiting miR‑125b‑5p to increase myocardial repair. The present study may provide a potential therapeutic approach to improve myocardial repair following AMI.
Collapse
Affiliation(s)
- Xin-Long Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yuan-Yuan Zhao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Li Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yu Shi
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zhu-Qian Li
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xiang-Dong Zhao
- Department of Clinical Laboratory, Zhenjiang Provincial Blood Center, Zhenjiang, Jiangsu 212000, P.R. China
| | - Chang-Gen Xu
- Department of Clinical Laboratory, Zhenjiang Provincial Blood Center, Zhenjiang, Jiangsu 212000, P.R. China
| | - Hong-Ge Ji
- Department of Clinical Laboratory, Zhenjiang Provincial Blood Center, Zhenjiang, Jiangsu 212000, P.R. China
| | - Mei Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wen-Rong Xu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Wei Zhu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
27
|
Varga ZV, Ágg B, Ferdinandy P. miR-125b is a protectomiR: A rising star for acute cardioprotection. J Mol Cell Cardiol 2017; 115:51-53. [PMID: 29294330 DOI: 10.1016/j.yjmcc.2017.12.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 12/27/2017] [Indexed: 11/15/2022]
Affiliation(s)
- Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Bence Ágg
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Heart and Vascular Center, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary.
| |
Collapse
|
28
|
Zeng J, Hu J, Lian Y, Jiang Y, Chen B. SFRP5 is a target gene transcriptionally regulated by PPARγ in 3T3-L1 adipocytes. Gene 2017; 641:190-195. [PMID: 29066306 DOI: 10.1016/j.gene.2017.10.066] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/16/2017] [Accepted: 10/20/2017] [Indexed: 12/12/2022]
Abstract
Secreted frizzled-related protein 5 (SFRP5) is a newly identified adipokine. SFRP5 expression increases during the differentiation and maturation of adipocytes, but the factors regulating SFRP5 expression during this process remain unclear. This study showed that peroxisome proliferator-activated receptor γ (PPARγ) adenovirus transfection could enhance the SFRP5 expression of 3T3-L1 adipocytes. Three potential binding sites of PPARγ in the SFRP5 promoter domain were found by bioinformatics analysis. Luciferase reporter gene assay demonstrated that PPARγ regulated the activity of the SFRP5 promoter through cis-acting elements at -2,284--1,500bp. Further experiments verified that PPARγ could specifically bind to the SFRP5 promoter at -2,284--2,263bp using chromatin immunoprecipitation and electrophoretic mobility shift assay. These results suggest that SFRP5 be a target gene of PPARγ, and its expression may be under the transcriptional regulation of PPARγ.
Collapse
Affiliation(s)
- Jun Zeng
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jiongyu Hu
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yu Lian
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Youzhao Jiang
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| | - Bing Chen
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|