151
|
Xu X, Zhang B, Wang Y, Shi S, Lv J, Fu Z, Gao X, Li Y, Wu H, Song Q. Renal fibrosis in type 2 cardiorenal syndrome: An update on mechanisms and therapeutic opportunities. Biomed Pharmacother 2023; 164:114901. [PMID: 37224755 DOI: 10.1016/j.biopha.2023.114901] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/26/2023] Open
Abstract
Cardiorenal syndrome (CRS) is a state of coexisting heart failure and renal insufficiency in which acute or chronic dysfunction of the heart or kidney lead to acute or chronic dysfunction of the other organ.It was found that renal fibrosis is an important pathological process in the progression of type 2 CRS to end-stage renal disease, and progressive renal impairment accelerates the deterioration of cardiac function and significantly increases the hospitalization and mortality rates of patients. Previous studies have found that Hemodynamic Aiteration, RAAS Overactivation, SNS Dysfunction, Endothelial Dysfunction and Imbalance of natriuretic peptide system contribute to the development of renal disease in the decompensated phase of heart failure, but the exact mechanisms is not clear. Therefore, in this review, we focus on the molecular pathways involved in the development of renal fibrosis due to heart failure and identify the canonical and non-canonical TGF-β signaling pathways and hypoxia-sensing pathways, oxidative stress, endoplasmic reticulum stress, pro-inflammatory cytokines and chemokines as important triggers and regulators of fibrosis development, and summarize the therapeutic approaches for the above signaling pathways, including SB-525334 Sfrp1, DKK1, IMC, rosarostat, 4-PBA, etc. In addition, some potential natural drugs for this disease are also summarized, including SQD4S2, Wogonin, Astragaloside, etc.
Collapse
Affiliation(s)
- Xia Xu
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yajiao Wang
- College of Traditional Chinese Medicine, China Academy of Chinese Medical Science, Beijing, China
| | - Shuqing Shi
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiayu Lv
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenyue Fu
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Xiya Gao
- College of Traditional Chinese Medicine, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Yumeng Li
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Huaqin Wu
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qingqiao Song
- Department of General Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
152
|
Pan KL, Hsu YC, Chang ST, Chung CM, Lin CL. The Role of Cardiac Fibrosis in Diabetic Cardiomyopathy: From Pathophysiology to Clinical Diagnostic Tools. Int J Mol Sci 2023; 24:ijms24108604. [PMID: 37239956 DOI: 10.3390/ijms24108604] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disorder characterized by hyperglycemia due to inadequate insulin secretion, resistance, or both. The cardiovascular complications of DM are the leading cause of morbidity and mortality in diabetic patients. There are three major types of pathophysiologic cardiac remodeling including coronary artery atherosclerosis, cardiac autonomic neuropathy, and DM cardiomyopathy in patients with DM. DM cardiomyopathy is a distinct cardiomyopathy characterized by myocardial dysfunction in the absence of coronary artery disease, hypertension, and valvular heart disease. Cardiac fibrosis, defined as the excessive deposition of extracellular matrix (ECM) proteins, is a hallmark of DM cardiomyopathy. The pathophysiology of cardiac fibrosis in DM cardiomyopathy is complex and involves multiple cellular and molecular mechanisms. Cardiac fibrosis contributes to the development of heart failure with preserved ejection fraction (HFpEF), which increases mortality and the incidence of hospitalizations. As medical technology advances, the severity of cardiac fibrosis in DM cardiomyopathy can be evaluated by non-invasive imaging modalities such as echocardiography, heart computed tomography (CT), cardiac magnetic resonance imaging (MRI), and nuclear imaging. In this review article, we will discuss the pathophysiology of cardiac fibrosis in DM cardiomyopathy, non-invasive imaging modalities to evaluate the severity of cardiac fibrosis, and therapeutic strategies for DM cardiomyopathy.
Collapse
Affiliation(s)
- Kuo-Li Pan
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi City 613, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City 333, Taiwan
- Heart Failure Center, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi City 613, Taiwan
| | - Yung-Chien Hsu
- Department of Nephrology, Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi Branch, Chiayi City 613, Taiwan
| | - Shih-Tai Chang
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi City 613, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City 333, Taiwan
| | - Chang-Min Chung
- Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chiayi Branch, Chiayi City 613, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City 333, Taiwan
| | - Chun-Liang Lin
- College of Medicine, Chang Gung University, Taoyuan City 333, Taiwan
- Department of Nephrology, Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi Branch, Chiayi City 613, Taiwan
- Kidney Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Department of Medical Research, Chang Gung Memorial Hospital, Kaohsiung City 833, Taiwan
| |
Collapse
|
153
|
Jian Y, Zhou X, Shan W, Chen C, Ge W, Cui J, Yi W, Sun Y. Crosstalk between macrophages and cardiac cells after myocardial infarction. Cell Commun Signal 2023; 21:109. [PMID: 37170235 PMCID: PMC10173491 DOI: 10.1186/s12964-023-01105-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 03/18/2023] [Indexed: 05/13/2023] Open
Abstract
Cardiovascular diseases, such as myocardial infarction (MI), are a leading cause of death worldwide. Acute MI (AMI) inflicts massive injury to the coronary microcirculation, causing large-scale cardiomyocyte death due to ischemia and hypoxia. Inflammatory cells such as monocytes and macrophages migrate to the damaged area to clear away dead cells post-MI. Macrophages are pleiotropic cells of the innate immune system, which play an essential role in the initial inflammatory response that occurs following MI, inducing subsequent damage and facilitating recovery. Besides their recognized role within the immune response, macrophages participate in crosstalk with other cells (including cardiomyocytes, fibroblasts, immune cells, and vascular endothelial cells) to coordinate post-MI processes within cardiac tissue. Macrophage-secreted exosomes have recently attracted increasing attention, which has led to a more elaborate understanding of macrophage function. Currently, the functional roles of macrophages in the microenvironment of the infarcted heart, particularly with regard to their interaction with surrounding cells, remain unclear. Understanding the specific mechanisms that mediate this crosstalk is essential in treating MI. In this review, we discuss the origin of macrophages, changes in their distribution post-MI, phenotypic and functional plasticity, as well as the specific signaling pathways involved, with a focus on the crosstalk with other cells in the heart. Thus, we provide a new perspective on the treatment of MI. Further in-depth research is required to elucidate the mechanisms underlying crosstalk between macrophages and other cells within cardiac tissue for the identification of potential therapeutic targets. Video Abstract.
Collapse
Affiliation(s)
- Yuhong Jian
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiao Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenju Shan
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Cheng Chen
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wei Ge
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Yang Sun
- Department of General Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
154
|
Uhlig M, Hein M, Habigt MA, Tolba RH, Braunschweig T, Helmedag MJ, Arici M, Theißen A, Klinkenberg A, Klinge U, Mechelinck M. Cirrhotic Cardiomyopathy Following Bile Duct Ligation in Rats-A Matter of Time? Int J Mol Sci 2023; 24:8147. [PMID: 37175858 PMCID: PMC10249007 DOI: 10.3390/ijms24098147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/28/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
Cirrhotic patients often suffer from cirrhotic cardiomyopathy (CCM). Previous animal models of CCM were inconsistent concerning the time and mechanism of injury; thus, the temporal dynamics and cardiac vulnerability were studied in more detail. Rats underwent bile duct ligation (BDL) and a second surgery 28 days later. Cardiac function was assessed by conductance catheter and echocardiography. Histology, gene expression, and serum parameters were analyzed. A chronotropic incompetence (Pd31 < 0.001) and impaired contractility at rest and a reduced contractile reserve (Pd31 = 0.03, Pdob-d31 < 0.001) were seen 31 days after BDL with increased creatine (Pd35, Pd42, and Pd56 < 0.05) and transaminases (Pd31 < 0.001). A total of 56 days after BDL, myocardial fibrosis was seen (Pd56 < 0.001) accompanied by macrophage infiltration (CD68: Pgroup < 0.001) and systemic inflammation (TNFα: Pgroup < 0.001, white blood cell count: Pgroup < 0.001). Myocardial expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) was increased after 31 (Pd31 < 0.001) and decreased after 42 (Pd42 < 0.001) and 56 days (Pd56 < 0.001). Caspase-3 expression was increased 31 and 56 days after BDL (Pd31 = 0.005; Pd56 = 0.005). Structural changes in the myocardium were seen after 8 weeks. After the second surgery (second hit), transient myocardial insufficiency with secondary organ dysfunction was seen, characterized by reduced contractility and contractile reserve.
Collapse
Affiliation(s)
- Moritz Uhlig
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.H.); (M.A.H.); (A.T.); (M.M.)
| | - Marc Hein
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.H.); (M.A.H.); (A.T.); (M.M.)
| | - Moriz A. Habigt
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.H.); (M.A.H.); (A.T.); (M.M.)
| | - René H. Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany;
| | - Till Braunschweig
- Department of Pathology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany;
| | - Marius J. Helmedag
- Department of General, Visceral and Transplantation Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.J.H.); (U.K.)
| | - Melissa Arici
- Luisenhospital, 52064 Aachen, Germany; (M.A.); (A.K.)
| | - Alexander Theißen
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.H.); (M.A.H.); (A.T.); (M.M.)
| | | | - Uwe Klinge
- Department of General, Visceral and Transplantation Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.J.H.); (U.K.)
| | - Mare Mechelinck
- Department of Anesthesiology, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany; (M.H.); (M.A.H.); (A.T.); (M.M.)
- Institute for Laboratory Animal Science and Experimental Surgery, Faculty of Medicine, RWTH Aachen University, 52074 Aachen, Germany;
| |
Collapse
|
155
|
Broughton K, Esquer C, Echeagaray O, Firouzi F, Shain G, Ebeid D, Monsanto M, Yaareb D, Golgolab L, Gude N, Sussman MA. Surface Lin28A expression consistent with cellular stress parallels indicators of senescence. Cardiovasc Res 2023; 119:743-758. [PMID: 35880724 PMCID: PMC10409908 DOI: 10.1093/cvr/cvac122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 06/03/2022] [Accepted: 06/26/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Declining cellular functional capacity resulting from stress or ageing is a primary contributor to impairment of myocardial performance. Molecular pathway regulation of biological processes in cardiac interstitial cells (CICs) is pivotal in stress and ageing responses. Altered localization of the RNA-binding protein Lin28A has been reported in response to environmental stress, but the role of Lin28A in response to stress in CICs has not been explored. Surface Lin28A redistribution is indicative of stress response in CIC associated with ageing and senescence. METHODS AND RESULTS Localization of Lin28A was assessed by multiple experimental analyses and treatment conditions and correlated to oxidative stress, senescence, and ploidy in adult murine CICs. Surface Lin28A expression is present on 5% of fresh CICs and maintained through Passage 2, increasing to 21% in hyperoxic conditions but lowered to 14% in physiologic normoxia. Surface Lin28A is coincident with elevated senescence marker p16 and beta-galactosidase (β-gal) expression in CICs expanded in hyperoxia, and also increases with polyploidization and binucleation of CICs regardless of oxygen culture. Transcriptional profiling of CICs using single-cell RNA-Seq reveals up-regulation of pathways associated with oxidative stress in CICs exhibiting surface Lin28A. Induction of surface Lin28A by oxidative stress is blunted by treatment of cells with the antioxidant Trolox in a dose-dependent manner, with 300 μM Trolox exposure maintaining characteristics of freshly isolated CICs possessing low expression of surface Lin28A and β-gal with predominantly diploid content. CONCLUSION Surface Lin28A is a marker of environmental oxidative stress in CICs and antioxidant treatment antagonizes this phenotype. The biological significance of Lin28 surface expression and consequences for myocardial responses may provide important insights regarding mitigation of cardiac stress and ageing.
Collapse
Affiliation(s)
- Kathleen Broughton
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Carolina Esquer
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Oscar Echeagaray
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Fareheh Firouzi
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Grant Shain
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - David Ebeid
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Megan Monsanto
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Dena Yaareb
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Leila Golgolab
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Natalie Gude
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Mark A Sussman
- San Diego State University Heart Institute and Department of Biology, San Diego State University, 5500 Campanile Drive, San Diego, CA 92182, USA
| |
Collapse
|
156
|
Chen Y, Peng D. New insights into the molecular mechanisms of SGLT2 inhibitors on ventricular remodeling. Int Immunopharmacol 2023; 118:110072. [PMID: 37018976 DOI: 10.1016/j.intimp.2023.110072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 03/09/2023] [Accepted: 03/20/2023] [Indexed: 04/05/2023]
Abstract
Ventricular remodeling is a pathological process of ventricular response to continuous stimuli such as pressure overload, ischemia or ischemia-reperfusion, which can lead to the change of cardiac structure and function structure, which is central to the pathophysiology of heart failure (HF) and is an established prognostic factor in patients with HF. Sodium glucose cotransporter 2 inhibitors (SGLT2i) get a new hypoglycemic drug that inhibit sodium glucose coconspirator on renal tubular epithelial cells. Recently, clinical trials increasingly and animal experiments increasingly have shown that SGLT2 inhibitors have been largely applied in the fields of cardiovascular diseases, forinstance heart failure, myocardial ischemia-reperfusion injury, myocardial infarction, atrial fibrillation, metabolic diseases such as obesity, diabetes cardiomyopathy and other diseases play a cardiovascular protective role in addition to hypoglycemic. These diseases are association with ventricular remodeling. Inhibiting ventricular remodeling can improve the readmission rate and mortality of patients with heart failure. So far, clinical trials and animal experiments demonstrate that the protective effect of SGLT2 inhibitors in the cardiovascular field is bound to inhibit ventricular remodeling. Therefore, this review briefly investigates the molecular mechanisms of SGLT2 inhibitors on ameliorating ventricular remodeling, and further explore the mechanisms of cardiovascular protection of SGLT2 inhibitors, in order to establish strategies for ventricular remodeling to prevent the progress of heart failure.
Collapse
|
157
|
Todorova VB, Baxan N, Delahaye M, Harding SE, Rankin SM. Drug-based mobilisation of mesenchymal stem/stromal cells improves cardiac function post myocardial infarction. Dis Model Mech 2023; 16:dmm049630. [PMID: 36263604 PMCID: PMC10655717 DOI: 10.1242/dmm.049630] [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: 05/04/2022] [Accepted: 09/14/2022] [Indexed: 11/20/2022] Open
Abstract
There is an unmet need for treatments that prevent the progressive cardiac dysfunction following myocardial infarction. Mesenchymal stem/stromal cells (MSCs) are under investigation for cardiac repair; however, culture expansion prior to transplantation is hindering their homing and reparative abilities. Pharmacological mobilisation could be an alternative to MSC transplantation. Here, we report that endogenous MSCs mobilise into the circulation at day 5 post myocardial infarction in male Lewis rats. This mobilisation can be significantly increased by using a combination of the FDA-approved drugs mirabegron (β3-adrenoceptor agonist) and AMD3100 (CXCR4 antagonist). Blinded cardiac magnetic resonance imaging analysis showed the treated group to have increased left ventricular ejection fraction and decreased end systolic volume at 5 weeks post myocardial infarction. The mobilised group had a significant decrease in plasma IL-6 and TNF-α levels, a decrease in interstitial fibrosis, and an increase in the border zone blood vessel density. Conditioned medium from blood-derived MSCs supported angiogenesis in vitro, as shown by tube formation and wound healing assays. Our data suggest a novel pharmacological strategy that enhances myocardial infarction-induced MSC mobilisation and improves cardiac function after myocardial infarction.
Collapse
Affiliation(s)
- Veneta B. Todorova
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Nicoleta Baxan
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Matthew Delahaye
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Sian E. Harding
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| | - Sara M. Rankin
- Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Myocardial Function, 72 Du Cane Road, London W12 0NN, UK
| |
Collapse
|
158
|
Wu C, Lin D, Ji J, Jiang Y, Jiang F, Wang Y. PCSK9 Inhibition Regulates Infarction-Induced Cardiac Myofibroblast Transdifferentiation via Notch1 Signaling. Cell Biochem Biophys 2023:10.1007/s12013-023-01136-1. [PMID: 37081375 DOI: 10.1007/s12013-023-01136-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 04/12/2023] [Indexed: 04/22/2023]
Abstract
Increasing evidence suggests that PCSK9 inhibition protects cardiomyocytes against ischemia-reperfusion injury after myocardial infarction. However, it is not clear whether PCSK9 inhibitor (PCSK9i) affects cardiac fibroblasts (CFs) activation after MI. In this study we used SBC-115076, an antagonist of PCSK9, to investigate the role of PCSK9i in the conversion of CFs to cardiac myofibroblasts (CMFs) after MI and provided a basic for its clinical application in cardiac fibrosis after MI. In vivo study, PCSK9i was injected into mice 4 days after MI. Cardiac function and degree of fibrosis were evaluated by echocardiographic and tissue staining after treatment. Western blot showed that PCSK9i treatment decreases expression of α-SMA, collagen and increases expression of Notch1 in border infarct area. Vitro studies showed that PCSK9i decreased the degree of fibrosis, migration, and collagen fiber deposition in CFs. Confocal microscopy imaging also showed that hypoxia contributes to the formation of α-SMA stress filaments, and PCSK9i alleviated this state. Moreover, overexpression of Notch1 further suppress the activation of CFs under hypoxia. These results revealed that SBC-115076 ameliorates cardiac fibrosis and ventricular dysfunction post-myocardial infarction through inhibition of the differentiation of cardiac fibroblasts to myofibroblasts via Notch1/Hes1 signaling.
Collapse
Affiliation(s)
- Chen Wu
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dawei Lin
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Ji
- Department of Cardiology, Chongming Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yiweng Jiang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Jiang
- Clinical Research & Innovation Unit, Chongming Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Yaosheng Wang
- Department of Cardiology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of Cardiology, Chongming Hospital Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai, China.
- Clinical Research & Innovation Unit, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
159
|
Leancă SA, Afrăsânie I, Crișu D, Matei IT, Duca ȘT, Costache AD, Onofrei V, Tudorancea I, Mitu O, Bădescu MC, Șerban LI, Costache II. Cardiac Reverse Remodeling in Ischemic Heart Disease with Novel Therapies for Heart Failure with Reduced Ejection Fraction. Life (Basel) 2023; 13:1000. [PMID: 37109529 PMCID: PMC10143569 DOI: 10.3390/life13041000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/29/2023] Open
Abstract
Despite the improvements in the treatment of coronary artery disease (CAD) and acute myocardial infarction (MI) over the past 20 years, ischemic heart disease (IHD) continues to be the most common cause of heart failure (HF). In clinical trials, over 70% of patients diagnosed with HF had IHD as the underlying cause. Furthermore, IHD predicts a worse outcome for patients with HF, leading to a substantial increase in late morbidity, mortality, and healthcare costs. In recent years, new pharmacological therapies have emerged for the treatment of HF, such as sodium-glucose cotransporter-2 inhibitors, angiotensin receptor-neprilysin inhibitors, selective cardiac myosin activators, and oral soluble guanylate cyclase stimulators, demonstrating clear or potential benefits in patients with HF with reduced ejection fraction. Interventional strategies such as cardiac resynchronization therapy, cardiac contractility modulation, or baroreflex activation therapy might provide additional therapeutic benefits by improving symptoms and promoting reverse remodeling. Furthermore, cardiac regenerative therapies such as stem cell transplantation could become a new therapeutic resource in the management of HF. By analyzing the existing data from the literature, this review aims to evaluate the impact of new HF therapies in patients with IHD in order to gain further insight into the best form of therapeutic management for this large proportion of HF patients.
Collapse
Affiliation(s)
- Sabina Andreea Leancă
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Afrăsânie
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Daniela Crișu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Iulian Theodor Matei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ștefania Teodora Duca
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alexandru Dan Costache
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Department of Cardiovascular Rehabilitation, Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Viviana Onofrei
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ionuţ Tudorancea
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Ovidiu Mitu
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Minerva Codruța Bădescu
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Internal Medicine Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Lăcrămioara Ionela Șerban
- Department of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Irina Iuliana Costache
- Cardiology Clinic, “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
- Department of Internal Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| |
Collapse
|
160
|
Ren C, Zhao X, Liu K, Wang L, Chen Q, Jiang H, Gao X, Lv X, Zhi X, Wu X, Li Y. Research progress of natural medicine Astragalus mongholicus Bunge in treatment of myocardial fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 305:116128. [PMID: 36623754 DOI: 10.1016/j.jep.2022.116128] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/25/2022] [Accepted: 12/30/2022] [Indexed: 06/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Myocardial fibrosis (MF) is a common pathological manifestation of many cardiovascular diseases at a certain stage, with excessive accumulation of collagen fibers, excessive increase in collagen content, and a significant increase in collagen volume as the main pathological changes. There are currently no effective drugs for the treatment of myocardial fibrosis. Traditional Chinese medicine (TCM), the main component of the medical practice used for more than 5000 years, especially in China, often exerts a wider action spectrum than previously attempted options in treating human diseases. In recent times, the great potential of TCM in the treatment of MF has received much attention. Especially many experimental studies on the treatment of MF by Astragalus mongholicus Bunge have been conducted, and the effect is remarkable, which may provide more comprehensive database and theoretical support for the application of Astragalus mongholicus Bunge in the treatment of MF and could be considered a promising candidate drug for preventing MF. AIM OF THE REVIEW This review summarizes the chemical components of Astragalus mongholicus Bunge, Astragalus mongholicus Bunge extract, Astragalus mongholicus Bunge single prescription, and Astragalus mongholicus Bunge compound preparation in the treatment of MF, and provides comprehensive information and a reliable basis for the exploration of new treatment strategies of botanical drugs in the therapy of MF. METHODS The literature information was obtained from the scientific databases on ethnobotany and ethnomedicines (up to August 2022), mainly from the PubMed, Web of Science, and CNKI databases. The experimental studies on the anti-myocardial fibrosis role of the effective active components of Astragalus mongholicus Bunge and the utility of its compound preparation and the involved mechanisms were identified. The search keywords for such work included: "myocardial fibrosis" or "Cardiac fibrosis ", and "Astragalus mongholicus Bunge", "extract," or "herb". RESULTS Several studies have shown that the effective active components of Astragalus mongholicus Bunge and its formulas, particularly Astragaloside IV, Astragalus polysaccharide, total saponins of Astragalus mongholicus Bunge, triterpenoid saponins of Astragalus mongholicus Bunge, and cycloastragenol, exhibit potential benefits against MF, the mechanisms of which appear to involve the regulation of inflammation, oxidant stress, and pro-fibrotic signaling pathways, etc. Conclusion: These research works have shown the therapeutic benefits of Astragalus mongholicus Bunge in the treatment of MF. However, further research should be undertaken to clarify the unconfirmed chemical composition and regulatory mechanisms, conduct standard clinical trials, and evaluate the possible side effects. The insights in the present review provided rich ideas for developing new anti-MF drugs. THESIS Myocardial fibrosis (MF) with excessive accumulation of collagen fibers, excessive increase in collagen content, and a significant increase in collagen volume as the main pathological changes is a common pathological manifestation of many cardiovascular diseases at a certain stage, which seriously affects cardiac function. At present, there is still a lack of effective drugs for the treatment of MF. Traditional Chinese medicine (TCM), the main component of the medical practice used for more than 5000 years especially in China, often exerts wider action spectrum than previously attempted options in treating human diseases. In recent times, the great potential of TCM in the treatment of MF has received much attention. Especially many experimental studies on the treatment of MF by Astragalus mongholicus Bunge have been conducted, and the effect is remarkable, which may provide more comprehensive data base and theoretical support for the application of Astragalus mongholicus Bunge in the treatment of MF and could be considered a promising candidate drug for preventing MF.
Collapse
Affiliation(s)
- Chunzhen Ren
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - Xinke Zhao
- Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Kai Liu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - Lirong Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - Qilin Chen
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - Hugang Jiang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China
| | - Xiang Gao
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xinfang Lv
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xiaodong Zhi
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China; Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xue Wu
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China; The second hospital of Lanzhou University, Lanzhou, 730000, China
| | - Yingdong Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Gansu Province Key Laboratory of Chinese Medicine for the Prevention andTreatment of Chronic Diseases, Lanzhou, 730000, China; Key clinical specialty of the National Health Commission of the People's Republic of China, Key Specialized Cardiovascular Laboratory National Administration of Traditional Chinese Medicine, Lanzhou, 730000, China.
| |
Collapse
|
161
|
McElhinney K, Irnaten M, O’Brien C. p53 and Myofibroblast Apoptosis in Organ Fibrosis. Int J Mol Sci 2023; 24:ijms24076737. [PMID: 37047710 PMCID: PMC10095465 DOI: 10.3390/ijms24076737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/23/2023] [Accepted: 03/28/2023] [Indexed: 04/07/2023] Open
Abstract
Organ fibrosis represents a dysregulated, maladaptive wound repair response that results in progressive disruption of normal tissue architecture leading to detrimental deterioration in physiological function, and significant morbidity/mortality. Fibrosis is thought to contribute to nearly 50% of all deaths in the Western world with current treatment modalities effective in slowing disease progression but not effective in restoring organ function or reversing fibrotic changes. When physiological wound repair is complete, myofibroblasts are programmed to undergo cell death and self-clearance, however, in fibrosis there is a characteristic absence of myofibroblast apoptosis. It has been shown that in fibrosis, myofibroblasts adopt an apoptotic-resistant, highly proliferative phenotype leading to persistent myofibroblast activation and perpetuation of the fibrotic disease process. Recently, this pathological adaptation has been linked to dysregulated expression of tumour suppressor gene p53. In this review, we discuss p53 dysregulation and apoptotic failure in myofibroblasts and demonstrate its consistent link to fibrotic disease development in all types of organ fibrosis. An enhanced understanding of the role of p53 dysregulation and myofibroblast apoptosis may aid in future novel therapeutic and/or diagnostic strategies in organ fibrosis.
Collapse
Affiliation(s)
- Kealan McElhinney
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | - Mustapha Irnaten
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| | - Colm O’Brien
- UCD Clinical Research Centre, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland
| |
Collapse
|
162
|
Shiraishi M, Suzuki K, Yamaguchi A. Effect of mechanical tension on fibroblast transcriptome profile and regulatory mechanisms of myocardial collagen turnover. FASEB J 2023; 37:e22841. [PMID: 36856975 DOI: 10.1096/fj.202201899r] [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: 11/14/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 03/02/2023]
Abstract
Excess deposition of extracellular matrix in the myocardium is a predictor of reduced left ventricular function. Although reducing the hemodynamic load is known to improve myocardial fibrosis, the mechanisms underlying the reversal of the fibrosis have not been elucidated. We focused on the elasticity of myocardial tissue, which is assumed to influence the fibroblast phenotype. Normal and fibrotic myocardium were cultured in 16 kPa and 64 kPa silicone gel-coated dishes supplemented with recombinant TGFβ protein, respectively. Matrix-degrading myocardium was cultured in 64 kPa silicone gel-coated dishes with recombinant TGFβ protein and then in 16 kPa silicone gel-coated dishes. Cardiac fibroblasts were cultured in this three-part in vitro pathological models and compared. Fibroblasts differentiated into activated or matrix-degrading types in response to the pericellular environment. Comprehensive gene expression analysis of fibroblasts in each in vitro condition showed Selenbp1 to be one of the genes responsible for regulating differentiation of fibroblasts. In vitro knockdown of Selenbp1 enhanced fibroblast activation and inhibited conversion to the matrix-degrading form. In vivo knockdown of Selenbp1 resulted in structural changes in the left ventricle associated with progressive tissue fibrosis and left ventricular diastolic failure. Selenbp1 is involved in regulating fibroblast differentiation and appears to be one of the major molecules regulating collagen turnover in cardiac fibrosis.
Collapse
Affiliation(s)
- Manabu Shiraishi
- Department of Cardiovascular Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| | - Ken Suzuki
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- National Cerebral and Cardiovascular Center Hospital, Osaka, Japan
| | - Atsushi Yamaguchi
- Department of Cardiovascular Surgery, Saitama Medical Center, Jichi Medical University, Saitama, Japan
| |
Collapse
|
163
|
Alrasheed NM, Alammari RB, Alshammari TK, Alamin MA, Alharbi AO, Alonazi AS, Bin Dayel AF, Alrasheed NM. α1A Adrenoreceptor blockade attenuates myocardial infarction by modulating the integrin-linked kinase/TGF-β/Smad signaling pathways. BMC Cardiovasc Disord 2023; 23:153. [PMID: 36964489 PMCID: PMC10037904 DOI: 10.1186/s12872-023-03188-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/16/2023] [Indexed: 03/26/2023] Open
Abstract
Background Myocardial infarction (MI) is considered a public health problem. According to the World Health Organization, MI is a leading cause of death and comorbidities worldwide. Activation of the α1A adrenergic receptor is a contributing factor to the development of MI. Tamsulosin, an α1A adrenergic blocker, has gained wide popularity as a medication for the treatment of benign prostatic hyperplasia. Limited evidence from previous studies has revealed the potential cardioprotective effects of tamsulosin, as its inhibitory effect on the α1A adrenoceptor protects the heart by acting on the smooth muscle of blood vessels, which results in hypotension; however, its effect on the infarcted heart is still unclear. The mechanisms of the expected cardioprotective effects mediated by tamsulosin are not yet understood. Transforming growth factor-beta (TGF-β), a mediator of fibrosis, is considered an attractive therapeutic target for remodeling after MI. The role of α1A adrenoceptor inhibition or its relationships with integrin-linked kinase (ILK) and TGF-β/small mothers against decapentaplegic (Smad) signaling pathways in attenuating MI are unclear. The present study was designed to investigate whether tamsulosin attenuates MI by modulating an ILK-related TGF-β/Smad pathway. Methods Twenty-four adult male Wistar rats were randomly divided into 4 groups: control, ISO, TAM, and ISO + TAM. ISO (150 mg/kg, intraperitoneally) was injected on Days 20 and 21 to induce MI. Tamsulosin (0.8 mg/kg, orally) was administered for 21 days, prior to ISO injection for 2 consecutive days. Heart-to-body weight ratios and cardiac and fibrotic biomarker levels were subsequently determined. ILK, TGF-β1, p-Smad2/3, and collagen III protein expression levels were determined using biomolecular methods. Results Tamsulosin significantly attenuated the relative heart-to-body weight index (p < 0.5) and creatine kinase-MB level (p < 0.01) compared with those in the ISO control group. While ISO resulted in superoxide anion production and enhanced oxidative damage, tamsulosin significantly prevented this damage through antioxidant defense mechanisms, increasing glutathione and superoxide dismutase levels (p < 0.05) and decreasing lipid peroxide oxidation levels (p < 0.01). The present data revealed that tamsulosin reduced TGF-β/p-Smad2/3 expression and enhanced ILK expression. Conclusion Tamsulosin may exert a cardioprotective effect by modulating the ILK-related TGF-β/Smad signaling pathway. Thus, tamsulosin may be a useful therapeutic approach for preventing MI. Supplementary Information The online version contains supplementary material available at 10.1186/s12872-023-03188-w.
Collapse
Affiliation(s)
- Nawal M. Alrasheed
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Raghad B. Alammari
- grid.56302.320000 0004 1773 5396Pharm D. Student, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Tahani K. Alshammari
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Maha A. Alamin
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Abeer O. Alharbi
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Asma S. Alonazi
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Anfal F. Bin Dayel
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| | - Nouf M. Alrasheed
- grid.56302.320000 0004 1773 5396Department of Pharmacology and Toxicology, College of Pharmacy , King Saud University, P.O. Box 70474, Riyadh, 11567 Saudi Arabia
| |
Collapse
|
164
|
Zhou J, Tian G, Quan Y, Kong Q, Huang F, Li J, Wu W, Tang Y, Zhou Z, Liu X. The long noncoding RNA THBS1-AS1 promotes cardiac fibroblast activation in cardiac fibrosis by regulating TGFBR1. JCI Insight 2023; 8:160745. [PMID: 36787190 PMCID: PMC10070117 DOI: 10.1172/jci.insight.160745] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Cardiac fibrosis is associated with an adverse prognosis in cardiovascular disease that results in a decreased cardiac compliance and, ultimately, heart failure. Recent studies have identified the role of long noncoding RNA (lncRNA) in cardiac fibrosis. However, the functions of many lncRNAs in cardiac fibrosis remain to be characterized. Through a whole-transcriptome sequencing and bioinformatics analysis on a mouse model of pressure overload-induced cardiac fibrosis, we screened a key lncRNA termed thrombospondin 1 antisense 1 (THBS1-AS1), which was positively associated with cardiac fibrosis. In vitro functional studies demonstrated that the silencing of THBS1-AS1 ameliorated TGF-β1 effects on cardiac fibroblast (CF) activation, and the overexpression of THBS1-AS1 displayed the opposite effect. A mechanistic study revealed that THBS1-AS1 could sponge miR-221/222 to regulate the expression of TGFBR1. Moreover, under TGF-β1 stimulation, the forced expression of miR-221/222 or the knockdown TGFBR1 significantly reversed the THBS1-AS1 overexpression induced by further CF activation. In vivo, specific knockdown of THBS1-AS1 in activated CFs significantly alleviated transverse aorta constriction-induced (TAC-induced) cardiac fibrosis in mice. Finally, we demonstrated that the human THBS1-AS1 can also affect the activation of CFs by regulating TGFBR1. In conclusion, this study reveals that lncRNA THBS1-AS1 is a potentially novel regulator of cardiac fibrosis and may serve as a target for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Junteng Zhou
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center
- Health Management Center, General Practice Medical Center, and
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Geer Tian
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center
| | - Yue Quan
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center
| | - Qihang Kong
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center
| | - Fangyang Huang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Junli Li
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center
| | - Wenchao Wu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center
| | - Yong Tang
- International Joint Research Centre on Purinergic Signaling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Acupuncture & Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine Solna, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
165
|
Xi T, Wang R, Pi D, Ouyang J, Yang J. The p53/miR-29a-3p axis mediates the antifibrotic effect of leonurine on angiotensin II-stimulated rat cardiac fibroblasts. Exp Cell Res 2023; 426:113556. [PMID: 36933858 DOI: 10.1016/j.yexcr.2023.113556] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/20/2023] [Accepted: 03/15/2023] [Indexed: 03/18/2023]
Abstract
Overactivation of cardiac fibroblasts (CFs) is one of the main causes of myocardial fibrosis (MF), and inhibition of CF activation is a crucial strategy for MF therapy. A previous study by our group demonstrated that leonurine (LE) effectively inhibits collagen synthesis and myofibroblast generation originated from CFs, and eventually mitigates the progression of MF (where miR-29a-3p is likely to be a vital mediator). However, the underlying mechanisms involved in this process remain unknown. Thus, the present study aimed to investigate the precise role of miR-29a-3p in LE-treated CFs, and to elucidate the pharmacological effects of LE on MF. Neonatal rat CFs were isolated and stimulated by angiotensin II (Ang II) to mimic the pathological process of MF in vitro. The results show that LE distinctly inhibits collagen synthesis, as well as the proliferation, differentiation and migration of CFs, all of which could be induced by Ang II. In addition, LE promotes apoptosis in CFs under Ang II stimulation. During this process, the down-regulated expressions of miR-29a-3p and p53 are partly restored by LE. Either knockdown of miR-29a-3p or inhibition of p53 by PFT-α (a p53 inhibitor) blocks the antifibrotic effect of LE. Notably, PFT-α suppresses miR-29a-3p levels in CFs under both normal and Ang II-treated conditions. Furthermore, ChIP analysis confirmed that p53 is bound to the promoter region of miR-29a-3p, and directly regulates its expression. Overall, our study demonstrates that LE upregulates p53 and miR-29a-3p expression, and subsequently inhibits CF overactivation, suggesting that the p53/miR-29a-3p axis may play a crucial role in mediating the antifibrotic effect of LE against MF.
Collapse
Affiliation(s)
- Tianlan Xi
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Ruiyu Wang
- Institute of Life Science, Chongqing Medical University, Chongqing, China
| | - Damao Pi
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing Ouyang
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China.
| | - Jiadan Yang
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
166
|
Nicolini G, Balzan S, Forini F. Activated fibroblasts in cardiac and cancer fibrosis: An overview of analogies and new potential therapeutic options. Life Sci 2023; 321:121575. [PMID: 36933828 DOI: 10.1016/j.lfs.2023.121575] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/06/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023]
Abstract
Heart disease and cancer are two major causes of morbidity and mortality in the industrialized countries, and their increasingly recognized connections are shifting the focus from single disease studies to an interdisciplinary approach. Fibroblast-mediated intercellular crosstalk is critically involved in the evolution of both pathologies. In healthy myocardium and in non-cancerous conditions, resident fibroblasts are the main cell source for synthesis of the extracellular matrix (ECM) and important sentinels of tissue integrity. In the setting of myocardial disease or cancer, quiescent fibroblasts activate, respectively, into myofibroblasts (myoFbs) and cancer-associated fibroblasts (CAFs), characterized by increased production of contractile proteins, and by a highly proliferative and secretory phenotype. Although the initial activation of myoFbs/CAFs is an adaptive process to repair the damaged tissue, massive deposition of ECM proteins leads to maladaptive cardiac or cancer fibrosis, a recognized marker of adverse outcome. A better understanding of the key mechanisms orchestrating fibroblast hyperactivity may help developing innovative therapeutic options to restrain myocardial or tumor stiffness and improve patient prognosis. Albeit still unappreciated, the dynamic transition of myocardial and tumor fibroblasts into myoFbs and CAFs shares several common triggers and signaling pathways relevant to TGF-β dependent cascade, metabolic reprogramming, mechanotransduction, secretory properties, and epigenetic regulation, which might lay the foundation for future antifibrotic intervention. Therefore, the aim of this review is to highlight emerging analogies in the molecular signature underlying myoFbs and CAFs activation with the purpose of identifying novel prognostic/diagnostic biomarkers, and to elucidate the potential of drug repositioning strategies to mitigate cardiac/cancer fibrosis.
Collapse
Affiliation(s)
| | - Silvana Balzan
- CNR Institute of Clinical Physiology, Via G.Moruzzi 1, 56124 Pisa, Italy
| | - Francesca Forini
- CNR Institute of Clinical Physiology, Via G.Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
167
|
Li Y, Xu Y, Li W, Guo J, Wan K, Wang J, Xu Z, Han Y, Sun J, Chen Y. Cardiac MRI to Predict Sudden Cardiac Death Risk in Dilated Cardiomyopathy. Radiology 2023; 307:e222552. [PMID: 36916890 DOI: 10.1148/radiol.222552] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Abstract
Background Sudden cardiac death (SCD) is one of the leading causes of death in individuals with nonischemic dilated cardiomyopathy (DCM). However, the risk stratification of SCD events remains challenging in clinical practice. Purpose To determine whether myocardial tissue characterization with cardiac MRI could be used to predict SCD events and to explore a SCD stratification algorithm in nonischemic DCM. Materials and Methods In this prospective single-center study, adults with nonischemic DCM who underwent cardiac MRI between June 2012 and August 2020 were enrolled. SCD-related events included SCD, appropriate implantable cardioverter-defibrillator shock, and resuscitation after cardiac arrest. Competing risk regression analysis and Kaplan-Meier analysis were performed to identify the association of myocardial tissue characterization with outcomes. Results Among the 858 participants (mean age, 48 years; age range, 18-83 years; 603 men), 70 (8%) participants experienced SCD-related events during a median follow-up of 33.0 months. In multivariable competing risk analysis, late gadolinium enhancement (LGE) (hazard ratio [HR], 1.87; 95% CI: 1.07, 3.27; P = .03), native T1 (per 10-msec increase: HR, 1.07; 95% CI: 1.04, 1.11; P < .001), and extracellular volume fraction (per 3% increase: HR, 1.26; 95% CI: 1.11, 1.44; P < .001) were independent predictors of SCD-related events after adjustment of systolic blood pressure, atrial fibrillation, and left ventricular ejection fraction. An SCD risk stratification category was developed with a combination of native T1 and LGE. Participants with a native T1 value 4 or more SDs above the mean (1382 msec) had the highest annual SCD-related events rate of 9.3%, and participants with a native T1 value 2 SDs below the mean (1292 msec) and negative LGE had the lowest rate of 0.6%. This category showed good prediction ability (C statistic = 0.74) and could be used to discriminate SCD risk and competing heart failure risk. Conclusion Myocardial tissue characteristics derived from cardiac MRI were independent predictors of sudden cardiac death (SCD)-related events in individuals with nonischemic dilated cardiomyopathy and could be used to stratify participants according to different SCD risk categories. Clinical trial registration no. ChiCTR1800017058 © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Sakuma in this issue.
Collapse
Affiliation(s)
- Yangjie Li
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| | - Yuanwei Xu
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| | - Weihao Li
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| | - Jiajun Guo
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| | - Ke Wan
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| | - Jie Wang
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| | - Ziqian Xu
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| | - Yuchi Han
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| | - Jiayu Sun
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| | - Yucheng Chen
- From the Departments of Cardiology (Y.L., Y.X., W.L., J.G., J.W., Z.X., Y.C.), Geriatrics (K.W.), and Radiology (J.S.), West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China; and Wexner Medical Center, College of Medicine, The Ohio State University, Columbus, Ohio (Y.H.)
| |
Collapse
|
168
|
Brojakowska A, Jackson CJ, Bisserier M, Khlgatian MK, Grano C, Blattnig SR, Zhang S, Fish KM, Chepurko V, Chepurko E, Gillespie V, Dai Y, Lee B, Garikipati VNS, Hadri L, Kishore R, Goukassian DA. Lifetime Evaluation of Left Ventricular Structure and Function in Male C57BL/6J Mice after Gamma and Space-Type Radiation Exposure. Int J Mol Sci 2023; 24:5451. [PMID: 36982525 PMCID: PMC10049327 DOI: 10.3390/ijms24065451] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/06/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
The lifetime effects of space irradiation (IR) on left ventricular (LV) function are unknown. The cardiac effects induced by space-type IR, specifically 5-ion simplified galactic cosmic ray simulation (simGCRsim), are yet to be discovered. Three-month-old, age-matched, male C57BL/6J mice were irradiated with 137Cs gamma (γ; 100, 200 cGy) and simGCRsim (50 and 100 cGy). LV function was assessed via transthoracic echocardiography at 14 and 28 days (early), and at 365, 440, and 660 (late) days post IR. We measured the endothelial function marker brain natriuretic peptide in plasma at three late timepoints. We assessed the mRNA expression of the genes involved in cardiac remodeling, fibrosis, inflammation, and calcium handling in LVs harvested at 660 days post IR. All IR groups had impaired global LV systolic function at 14, 28, and 365 days. At 660 days, 50 cGy simGCRsim-IR mice exhibited preserved LV systolic function with altered LV size and mass. At this timepoint, the simGCRsim-IR mice had elevated levels of cardiac fibrosis, inflammation, and hypertrophy markers Tgfβ1, Mcp1, Mmp9, and βmhc, suggesting that space-type IR may induce the cardiac remodeling processes that are commonly associated with diastolic dysfunction. IR groups showing statistical significance were modeled to calculate the Relative Biological Effectiveness (RBE) and Radiation Effects Ratio (RER). The observed dose-response shape did not indicate a lower threshold at these IR doses. A single full-body IR at doses of 100-200 cGy for γ-IR, and 50-100 cGy for simGCRsim-IR decreases the global LV systolic function in WT mice as early as 14 and 28 days after exposure, and at 660 days post IR. Interestingly, there is an intermediate time point (365 days) where the impairment in LV function is observed. These findings do not exclude the possibility of increased acute or degenerative cardiovascular disease risks at lower doses of space-type IR, and/or when combined with other space travel-associated stressors such as microgravity.
Collapse
Affiliation(s)
- Agnieszka Brojakowska
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - Malik Bisserier
- Department of Cell Biology and Anatomy and Physiology, New York Medical College, Valhalla, NY 10595, USA
| | - Mary K. Khlgatian
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Cynthia Grano
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Steve R. Blattnig
- National Aeronautics and Space Administration, Hampton, VA 23669, USA
| | - Shihong Zhang
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kenneth M. Fish
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Vadim Chepurko
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elena Chepurko
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Virginia Gillespie
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ying Dai
- Center for Comparative Medicine and Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brooke Lee
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Venkata Naga Srikanth Garikipati
- Department of Emergency Medicine, Dorothy M. Davis Heart Lung and Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Lahouaria Hadri
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Center of Excellence for Translational Medicine and Pharmacology, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Raj Kishore
- Department of Cardiovascular Sciences, Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - David A. Goukassian
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
169
|
Nguyen JP, Ramirez-Sanchez I, Garate-Carrillo A, Navarrete-Yañez V, Carballo-Castañeda RA, Ceballos G, Moreno-Ulloa A, Villarreal F. Effects of aging and type 2 diabetes on cardiac structure and function: Underlying mechanisms. Exp Gerontol 2023; 173:112108. [PMID: 36708752 DOI: 10.1016/j.exger.2023.112108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/20/2023] [Accepted: 01/23/2023] [Indexed: 01/26/2023]
Abstract
We characterized long-term changes in cardiac structure and function in a high-fat diet/streptozotocin mouse model of aging and type 2 diabetes mellitus (T2D) and examined how the intersection of both conditions alters plasma metabolomics. We also evaluated the possible roles played by oxidative stress, arginase activity and pro-inflammatory cytokines. C57BL/6 male mice (13-month-old) were used. Control animals (n = 13) were fed regular chow for 10 months (aged group). T2D animals (n = 25) were provided a single injection of streptozotocin and fed a high fat diet for 10 months. In select endpoints, young animals were used for comparison. To monitor changes in left ventricular (LV) structure and function, echocardiography was used. At the terminal study (23 months), blood was collected and hearts processed for biochemical or histological analysis. Echo yielded diminished diastolic function with aging and T2D. LV fractional shortening and ejection fraction decreased with T2D by 16 months peaking at 23 months. Western blots noted increases in fibronectin and type I collagen with aging/T2D and greater levels with T2D in α-smooth muscle actin. Increases in plasma and/or myocardial protein carbonyls, arginase activity and pro-inflammatory cytokines occurred with aging and T2D. Untargeted metabolomics and cheminformatics revealed differences in the plasma metabolome of T2D vs. aged mice while select classes of lipid metabolites linked to insulin resistance, were dysregulated. We thus, document changes in LV structure and function with aging that in select endpoints, are accentuated with T2D and link them to increases in OS, arginase activity and pro-inflammatory cytokines.
Collapse
Affiliation(s)
| | - Israel Ramirez-Sanchez
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Alejandra Garate-Carrillo
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Viridiana Navarrete-Yañez
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | | | - Guillermo Ceballos
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Aldo Moreno-Ulloa
- Laboratorio MS2, Departamento de Innovación Biomédica, CICESE, Mexico
| | - Francisco Villarreal
- Veteran Affairs San Diego Health Care, San Diego, CA, USA; Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
170
|
Maroney SA, Siebert AE, Martinez ND, Rasmussen M, Peterson JA, Weiler H, Lincoln J, Mast AE. Platelet tissue factor pathway inhibitor-α dampens cardiac thrombosis and associated fibrosis in mice. J Thromb Haemost 2023; 21:639-651. [PMID: 36696221 PMCID: PMC10200073 DOI: 10.1016/j.jtha.2022.11.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/17/2022] [Accepted: 11/28/2022] [Indexed: 01/21/2023]
Abstract
BACKGROUND Tissue factor pathway inhibitor (TFPI) is the primary inhibitor of events initiating the blood coagulation pathway. Tfpi-/- mice die during embryonic development. The absence of protease-activated receptor (PAR) 4, the major thrombin receptor on mouse platelets, rescues Tfpi-/-mice to adulthood. Among the 3 TFPI isoforms in mice, TFPIα is the only isoform within platelets (pltTFPIα) and the only isoform that inhibits prothrombinase, the enzymatic complex that converts prothrombin to thrombin. OBJECTIVES To determine biological functions of pltTFPIα. METHODS Tfpi-/-/Par4-/- mice were irradiated and transplanted with bone marrow from mice lacking or containing pltTFPIα. Thus, PAR4 expression was restored in the recipient mice, which differed selectively by the presence or absence of pltTFPIα and lacked other forms of TFPI. RESULTS Recipient mice lacking pltTFPIα had reduced survival over the 200-day posttransplant period. Necropsy revealed radiation injury associated with large intraventricular platelet-rich thrombi, whereas other organs were not affected. Thrombi were associated with fibrotic presentations, including increased collagen deposition, periostin-positive activated fibroblasts, myofibroblasts, and macrophage infiltrates. Recipient mice containing pltTFPIα showed evidence of radiation injury but lacked heart pathology. CONCLUSIONS Tfpi-/-/Par4-/- mice develop severe cardiac fibrosis following irradiation and transplantation with bone marrow lacking pltTFPIα. This pathology is markedly reduced when the mice are transplanted with bone marrow containing pltTFPIα. Thus, in this model system pltTFPIα has an important physiological role in dampening pathological responses mediated by activated platelets within the heart tissue.
Collapse
Affiliation(s)
- Susan A Maroney
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Amy E Siebert
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Nicholas D Martinez
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Mark Rasmussen
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Julie A Peterson
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA
| | - Hartmut Weiler
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Joy Lincoln
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA; Division of Pediatric Cardiology, The Herma Heart Institute, Children's Wisconsin, Milwaukee, WI, USA
| | - Alan E Mast
- Thrombosis and Hemostasis Program, Versiti Blood Research Institute, Milwaukee, WI, USA; Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
171
|
Wang A, Li Z, Sun Z, Liu Y, Zhang D, Ma X. Potential Mechanisms Between HF and COPD: New Insights From Bioinformatics. Curr Probl Cardiol 2023; 48:101539. [PMID: 36528207 DOI: 10.1016/j.cpcardiol.2022.101539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Heart failure (HF) and chronic obstructive pulmonary disease (COPD) are closely related in clinical practice. This study aimed to investigate the co-genetic characteristics and potential molecular mechanisms of HF and COPD. HF and COPD datasets were downloaded from gene expression omnibus database. After identifying common differentially expressed genes (DEGs), the functional analysis highlighted the critical role of extracellular matrix and ribosomal signaling pathways in both diseases. In addition, GeneMANIA's results suggested that the 2 diseases were related to immune infiltration, and CIBERSORT suggested the role of macrophages. We also discovered 4 TFs and 1408 miRNAs linked to both diseases, and salbutamol may positively affect them.
Collapse
Affiliation(s)
- Anzhu Wang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhendong Li
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Zhuo Sun
- Qingdao West Coast New Area People's Hospital, Qingdao, China
| | - Yicheng Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dawu Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Xiaochang Ma
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China; National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China.
| |
Collapse
|
172
|
Fu H, Kong B, Zhu J, Huang H, Shuai W. Phenylacetylglutamine increases the susceptibility of ventricular arrhythmias in heart failure mice by exacerbated activation of the TLR4/AKT/mTOR signaling pathway. Int Immunopharmacol 2023; 116:109795. [PMID: 36736224 DOI: 10.1016/j.intimp.2023.109795] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/18/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023]
Abstract
BACKGROUND Intestinal microbial metabolites are a risk factor for cardiovascular diseases, and phenylacetylglutamine (PAGln) is a newly discovered intestinal metabolite in the latest study. In addition, elevated plasma PAGln concentration was associated with increased mortality and hospitalization rates in patients with heart failure (HF). However, the mechanism of PAGln leading to increased HF mortality is unclear. The present study was performed to investigate whether the PAGln deteriorated the susceptibility of ventricular arrhythmias (VAs) in the setting of HF. METHODS Thoracic aortic coarctation (TAC) was used to construct an animal model of HF in mice. Intraperitoneal injection of PAGln for 4 weeks intervened in HF mice. The concentration of PAGln was quantitatively determined by liquid chromatography-tandem mass spectrometry. Cardiac function was assessed by echocardiography; assessment of cardiac electrophysiological indexes was measured by electrocardiogram (ECG) and programmed electrical stimulation in isolated cardiac perfusion. Masson was stained for collagen deposition, and wheat germ agglutinin (WGA) was stained for the cross-sectional area of the myocytes. The qRT-PCR and Western Blotting were used to determine target gene expression in vivo and in vitro. RESULTS PAGln promoted the activation of cardiac inflammation and fibrosis and deteriorated cardiac function in HF mice. Moreover, PAGln extended APD90, shortened the ERP/APD90 and increased the incidence of VAs following HF in isolated heart perfusion. Mechanistically, PAGln significantly enhanced the activation of the TLR4/AKT/mTOR signaling pathway in vivo and in vitro. CONCLUSIONS PAGln increased the susceptibility of VAs in HF mice by activating the TLR4/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hui Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - Bin Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - Jun Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China
| | - He Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China.
| | - Wei Shuai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei, China; Cardiovascular Research Institute of Wuhan University, Wuhan 430060, Hubei, China; Hubei Key Laboratory of Cardiology, Wuhan 430060, Hubei, China.
| |
Collapse
|
173
|
Li J, Wang Y, Wang L, Qu Y, Zhou W, Zhong H, Xi D, Tang N, He F. Low expression of miR-1929-3p mediates murine cytomegalovirus-induced fibrosis in cardiac fibroblasts via targeting endothelin a receptor/NLRP3 inflammasome pathway. In Vitro Cell Dev Biol Anim 2023; 59:179-192. [PMID: 37002490 DOI: 10.1007/s11626-022-00742-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 11/22/2022] [Indexed: 04/03/2023]
Abstract
MicroRNAs are crucial in the development of myocardial remodeling in hypertension. Low miR-1929-3p expression induced by murine cytomegalovirus (MCMV) infection is closely related to hypertensive myocardial remodeling. This study investigated the molecular mechanism of miR-1929-3p-induced myocardial remodeling after MCMV infection. We modeled MCMV-infected mouse cardiac fibroblasts (MMCFs) as the primary cell model. First, MCMV infection reduced the expression of miR-1929-3p and increased the mRNA and protein expression of its target gene endothelin receptor type A (ETAR) in mouse cardiac fibroblasts (MCFs), which demonstrated an internal relationship with myocardial fibrosis (MF) based on high proliferation, phenotypic transformation (α-SMA), and collagen expression in MMCFs. The transfection of the miR-1929-3p mimic downregulated the high expression of ETAR and alleviated these adverse effects in MMCFs. Inversely, these effects were exacerbated by the miR-1929-3p inhibitor. Second, the transfection of endothelin receptor type A over-expressed adenovirus (adETAR) reversed these positive effects of the miR-1929-3p mimic on MF improvement. Third, the transfection of adETAR exhibited a strong inflammatory response in MMCFs with increased expression of NOD-like receptors pyrin domain containing 3 (NLRP3) and increased secretion of interleukin-18. However, we found that the ETAR antagonist BQ123 and the selected NLRP3 inflammasome inhibitor MCC950 effectively eliminated the inflammatory response induced by both MCMV infection and miR-1929-3p inhibitor. Moreover, the MCF supernatant was related to cardiomyocyte hypertrophy. Our findings suggest that MCMV infection promotes MF by inducing the downregulation of miR-1929-3p and the high expression of ETAR, which activates NLRP3 inflammasomes in MCFs.
Collapse
Affiliation(s)
- Jiayi Li
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - Yongjia Wang
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - LaMei Wang
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - Yuanyuan Qu
- Department of Respiratory Medicine, the First Affiliated Hospital of Medical College of Shihezi University, Shihezi, China
| | - Wei Zhou
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - Hua Zhong
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - DongMei Xi
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China
| | - Na Tang
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China.
| | - Fang He
- Department of Pathophysiology, Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, China.
| |
Collapse
|
174
|
Nassal DM, Shaheen R, Patel NJ, Yu J, Leahy N, Bibidakis D, Parinandi NL, Hund TJ. Spectrin-Based Regulation of Cardiac Fibroblast Cell-Cell Communication. Cells 2023; 12:748. [PMID: 36899883 PMCID: PMC10001335 DOI: 10.3390/cells12050748] [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: 11/04/2022] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Cardiac fibroblasts (CFs) maintain the fibrous extracellular matrix (ECM) that supports proper cardiac function. Cardiac injury induces a transition in the activity of CFs to promote cardiac fibrosis. CFs play a critical role in sensing local injury signals and coordinating the organ level response through paracrine communication to distal cells. However, the mechanisms by which CFs engage cell-cell communication networks in response to stress remain unknown. We tested a role for the action-associated cytoskeletal protein βIV-spectrin in regulating CF paracrine signaling. Conditioned culture media (CCM) was collected from WT and βIV-spectrin deficient (qv4J) CFs. WT CFs treated with qv4J CCM showed increased proliferation and collagen gel compaction compared to control. Consistent with the functional measurements, qv4J CCM contained higher levels of pro-inflammatory and pro-fibrotic cytokines and increased concentration of small extracellular vesicles (30-150 nm diameter, exosomes). Treatment of WT CFs with exosomes isolated from qv4J CCM induced a similar phenotypic change as that observed with complete CCM. Treatment of qv4J CFs with an inhibitor of the βIV-spectrin-associated transcription factor, STAT3, decreased the levels of both cytokines and exosomes in conditioned media. This study expands the role of the βIV-spectrin/STAT3 complex in stress-induced regulation of CF paracrine signaling.
Collapse
Affiliation(s)
- Drew M. Nassal
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Rebecca Shaheen
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Nehal J. Patel
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Jane Yu
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Nick Leahy
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Dimitra Bibidakis
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Narasimham L. Parinandi
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Thomas J. Hund
- The Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Medical Center, Columbus, OH 43210, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, Division of Cardiovascular Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
175
|
Duangrat R, Parichatikanond W, Likitnukul S, Mangmool S. Endothelin-1 Induces Cell Proliferation and Myofibroblast Differentiation through the ET AR/G αq/ERK Signaling Pathway in Human Cardiac Fibroblasts. Int J Mol Sci 2023; 24:ijms24054475. [PMID: 36901906 PMCID: PMC10002923 DOI: 10.3390/ijms24054475] [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: 01/16/2023] [Revised: 02/10/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Endothelin-1 (ET-1) has been implicated in the pathogenesis of cardiac fibrosis. Stimulation of endothelin receptors (ETR) with ET-1 leads to fibroblast activation and myofibroblast differentiation, which is mainly characterized by an overexpression of α-smooth muscle actin (α-SMA) and collagens. Although ET-1 is a potent profibrotic mediator, the signal transductions and subtype specificity of ETR contributing to cell proliferation, as well as α-SMA and collagen I synthesis in human cardiac fibroblasts are not well clarified. This study aimed to evaluate the subtype specificity and signal transduction of ETR on fibroblast activation and myofibroblast differentiation. Treatment with ET-1 induced fibroblast proliferation, and synthesis of myofibroblast markers, α-SMA, and collagen I through the ETAR subtype. Inhibition of Gαq protein, not Gαi or Gβγ, inhibited these effects of ET-1, indicating the essential role of Gαq protein-mediated ETAR signaling. In addition, ERK1/2 was required for ETAR/Gαq axis-induced proliferative capacity and overexpression of these myofibroblast markers. Antagonism of ETR with ETR antagonists (ERAs), ambrisentan and bosentan, inhibited ET-1-induced cell proliferation and synthesis of α-SMA and collagen I. Furthermore, ambrisentan and bosentan promoted the reversal of myofibroblasts after day 3 of treatment, with loss of proliferative ability and a reduction in α-SMA synthesis, confirming the restorative effects of ERAs. This novel work reports on the ETAR/Gαq/ERK signaling pathway for ET-1 actions and blockade of ETR signaling with ERAs, representing a promising therapeutic strategy for prevention and restoration of ET-1-induced cardiac fibrosis.
Collapse
Affiliation(s)
- Ratchanee Duangrat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Molecular Medicine Graduate Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Warisara Parichatikanond
- Department of Pharmacology, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
- Centre of Biopharmaceutical Science for Healthy Ageing (BSHA), Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Sutharinee Likitnukul
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Supachoke Mangmool
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Correspondence:
| |
Collapse
|
176
|
Sudi S, Thomas FM, Daud SK, Ag Daud DM, Sunggip C. The Pleiotropic Role of Extracellular ATP in Myocardial Remodelling. Molecules 2023; 28:molecules28052102. [PMID: 36903347 PMCID: PMC10004151 DOI: 10.3390/molecules28052102] [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: 01/26/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 03/12/2023] Open
Abstract
Myocardial remodelling is a molecular, cellular, and interstitial adaptation of the heart in response to altered environmental demands. The heart undergoes reversible physiological remodelling in response to changes in mechanical loading or irreversible pathological remodelling induced by neurohumoral factors and chronic stress, leading to heart failure. Adenosine triphosphate (ATP) is one of the potent mediators in cardiovascular signalling that act on the ligand-gated (P2X) and G-protein-coupled (P2Y) purinoceptors via the autocrine or paracrine manners. These activations mediate numerous intracellular communications by modulating the production of other messengers, including calcium, growth factors, cytokines, and nitric oxide. ATP is known to play a pleiotropic role in cardiovascular pathophysiology, making it a reliable biomarker for cardiac protection. This review outlines the sources of ATP released under physiological and pathological stress and its cell-specific mechanism of action. We further highlight a series of cardiovascular cell-to-cell communications of extracellular ATP signalling cascades in cardiac remodelling, which can be seen in hypertension, ischemia/reperfusion injury, fibrosis, hypertrophy, and atrophy. Finally, we summarize current pharmacological intervention using the ATP network as a target for cardiac protection. A better understanding of ATP communication in myocardial remodelling could be worthwhile for future drug development and repurposing and the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Suhaini Sudi
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Fiona Macniesia Thomas
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Siti Kadzirah Daud
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Dayang Maryama Ag Daud
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
- Health through Exercise and Active Living (HEAL) Research Unit, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Caroline Sunggip
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
- Borneo Medical and Health Research Centre, Faculty of Medicine and Health Sciences, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
- Correspondence:
| |
Collapse
|
177
|
Reproducing extracellular matrix adverse remodelling of non-ST myocardial infarction in a large animal model. Nat Commun 2023; 14:995. [PMID: 36813782 PMCID: PMC9945840 DOI: 10.1038/s41467-023-36350-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 01/23/2023] [Indexed: 02/24/2023] Open
Abstract
The rising incidence of non-ST-segment elevation myocardial infarction (NSTEMI) and associated long-term high mortality constitutes an urgent clinical issue. Unfortunately, the study of possible interventions to treat this pathology lacks a reproducible pre-clinical model. Indeed, currently adopted small and large animal models of MI mimic only full-thickness, ST-segment-elevation (STEMI) infarcts, and hence cater only for an investigation into therapeutics and interventions directed at this subset of MI. Thus, we develop an ovine model of NSTEMI by ligating the myocardial muscle at precise intervals parallel to the left anterior descending coronary artery. Upon histological and functional investigation to validate the proposed model and comparison with STEMI full ligation model, RNA-seq and proteomics show the distinctive features of post-NSTEMI tissue remodelling. Transcriptome and proteome-derived pathway analyses at acute (7 days) and late (28 days) post-NSTEMI pinpoint specific alterations in cardiac post-ischaemic extracellular matrix. Together with the rise of well-known markers of inflammation and fibrosis, NSTEMI ischaemic regions show distinctive patterns of complex galactosylated and sialylated N-glycans in cellular membranes and extracellular matrix. Identifying such changes in molecular moieties accessible to infusible and intra-myocardial injectable drugs sheds light on developing targeted pharmacological solutions to contrast adverse fibrotic remodelling.
Collapse
|
178
|
Du L, Sun X, Gong H, Wang T, Jiang L, Huang C, Xu X, Li Z, Xu H, Ma L, Li W, Chen T, Xu Q. Single cell and lineage tracing studies reveal the impact of CD34 + cells on myocardial fibrosis during heart failure. Stem Cell Res Ther 2023; 14:33. [PMID: 36805782 PMCID: PMC9942332 DOI: 10.1186/s13287-023-03256-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND CD34+ cells have been used to treat the patients with heart failure, but the outcome is variable. It is of great significance to scrutinize the fate and the mechanism of CD34+ cell differentiation in vivo during heart failure and explore its intervention strategy. METHODS We performed single-cell RNA sequencing (scRNA-seq) of the total non-cardiomyocytes and enriched Cd34-tdTomato+ lineage cells in the murine (male Cd34-CreERT2; Rosa26-tdTomato mice) pressure overload model (transverse aortic constriction, TAC), and total non-cardiomyocytes from human adult hearts. Then, in order to determine the origin of CD34+ cell that plays a role in myocardial fibrosis, bone marrow transplantation model was performed. Furthermore, to further clarify the role of CD34 + cells in myocardial remodeling in response to TAC injury, we generated Cd34-CreERT2; Rosa26-eGFP-DTA (Cre/DTA) mice. RESULTS By analyzing the transcriptomes of 59,505 single cells from the mouse heart and 22,537 single cells from the human heart, we illustrated the dynamics of cell landscape during the progression of heart hypertrophy, including CD34+ cells, fibroblasts, endothelial and immune cells. By combining genetic lineage tracing and bone marrow transplantation models, we demonstrated that non-bone-marrow-derived CD34+ cells give rise to fibroblasts and endothelial cells, while bone-marrow-derived CD34+ cell turned into immune cells only in response to pressure overload. Interestingly, partial depletion of CD34+ cells alleviated the severity of myocardial fibrosis with a significant improvement of cardiac function in Cd34-CreERT2; Rosa26-eGFP-DTA model. Similar changes of non-cardiomyocyte composition and cellular heterogeneity of heart failure were also observed in human patient with heart failure. Furthermore, immunostaining showed a double labeling of CD34 and fibroblast markers in human heart tissue. Mechanistically, our single-cell pseudotime analysis of scRNA-seq data and in vitro cell culture study revealed that Wnt-β-catenin and TGFβ1/Smad pathways are critical in regulating CD34+ cell differentiation toward fibroblasts. CONCLUSIONS Our study provides a cellular landscape of CD34+ cell-derived cells in the hypertrophy heart of human and animal models, indicating that non-bone-marrow-derived CD34+ cells differentiating into fibroblasts largely account for cardiac fibrosis. These findings may provide novel insights for the pathogenesis of cardiac fibrosis and have further potential therapeutic implications for the heart failure.
Collapse
Affiliation(s)
- Luping Du
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Xiaotong Sun
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Hui Gong
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Ting Wang
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Liujun Jiang
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Chengchen Huang
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Xiaodong Xu
- grid.452661.20000 0004 1803 6319Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Zhoubin Li
- grid.13402.340000 0004 1759 700XDepartment of Lung Transplantation, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Hongfei Xu
- grid.13402.340000 0004 1759 700XDepartment of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Liang Ma
- grid.13402.340000 0004 1759 700XDepartment of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China
| | - Weidong Li
- Department of Cardiovascular Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| | - Ting Chen
- Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China. .,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China.
| | - Qingbo Xu
- Department of Cardiology, School of Medicine, The First Affiliated Hospital, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003, Zhejiang, China.
| |
Collapse
|
179
|
Waldrop TI, Graham C, Gard W, Ingle K, Ptacek T, Nguyen N, Lose B, Sethu P, Lee T. Biomimetic cardiac tissue chip and murine arteriovenous fistula models for recapitulating clinically relevant cardiac remodeling under volume overload conditions. Front Bioeng Biotechnol 2023; 11:1101622. [PMID: 36873372 PMCID: PMC9978753 DOI: 10.3389/fbioe.2023.1101622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/03/2023] [Indexed: 02/18/2023] Open
Abstract
Cardiovascular events are the primary cause of death among dialysis patients. While arteriovenous fistulas (AVFs) are the access of choice for hemodialysis patients, AVF creation can lead to a volume overload (VO) state in the heart. We developed a three-dimensional (3D) cardiac tissue chip (CTC) with tunable pressure and stretch to model the acute hemodynamic changes associated with AVF creation to complement our murine AVF model of VO. In this study, we aimed to replicate the hemodynamics of murine AVF models in vitro and hypothesized that if 3D cardiac tissue constructs were subjected to "volume overload" conditions, they would display fibrosis and key gene expression changes seen in AVF mice. Mice underwent either an AVF or sham procedure and were sacrificed at 28 days. Cardiac tissue constructs composed of h9c2 rat cardiac myoblasts and normal adult human dermal fibroblasts in hydrogel were seeded into devices and exposed to 100 mg/10 mmHg pressure (0.4 s/0.6 s) at 1 Hz for 96 h. Controls were exposed to "normal" stretch and experimental group exposed to "volume overload". RT-PCR and histology were performed on the tissue constructs and mice left ventricles (LVs), and transcriptomics of mice LVs were also performed. Our tissue constructs and mice LV both demonstrated cardiac fibrosis as compared to control tissue constructs and sham-operated mice, respectively. Gene expression studies in our tissue constructs and mice LV demonstrated increased expression of genes associated with extracellular matrix production, oxidative stress, inflammation, and fibrosis in the VO conditions vs. control conditions. Our transcriptomics studies demonstrated activated upstream regulators related to fibrosis, inflammation, and oxidative stress such as collagen type 1 complex, TGFB1, CCR2, and VEGFA and inactivated regulators related to mitochondrial biogenesis in LV from mice AVF. In summary, our CTC model yields similar fibrosis-related histology and gene expression profiles as our murine AVF model. Thus, the CTC could potentially play a critical role in understanding cardiac pathobiology of VO states similar to what is present after AVF creation and may prove useful in evaluating therapies.
Collapse
Affiliation(s)
- Tatyana Isayeva Waldrop
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Caleb Graham
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - William Gard
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kevin Ingle
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Travis Ptacek
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Nguyen Nguyen
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bailey Lose
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Palaniappan Sethu
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Timmy Lee
- Department of Medicine and Division of Nephrology, University of Alabama at Birmingham, Birmingham, AL, United States
- Veterans Affairs Medical Center, Birmingham, AL, United States
| |
Collapse
|
180
|
Deng G, Ren J, Li R, Li M, Jin X, Li J, Liu J, Gao Y, Zhang J, Wang X, Wang G. Systematic investigation of the underlying mechanisms of GLP-1 receptor agonists to prevent myocardial infarction in patients with type 2 diabetes mellitus using network pharmacology. Front Pharmacol 2023; 14:1125753. [PMID: 36865917 PMCID: PMC9971732 DOI: 10.3389/fphar.2023.1125753] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/01/2023] [Indexed: 02/16/2023] Open
Abstract
Background: Several clinical trials have demonstrated that glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1RAs) reduce the incidence of non-fatal myocardial infarction (MI) in patients with type 2 diabetes mellitus (T2DM). However, the underlying mechanism remains unclear. In this study, we applied a network pharmacology method to investigate the mechanisms by which GLP-1RAs reduce MI occurrence in patients with T2DM. Methods: Targets of three GLP-1RAs (liraglutide, semaglutide, and albiglutide), T2DM, and MI were retrieved from online databases. The intersection process and associated targets retrieval were employed to obtain the related targets of GLP-1RAs against T2DM and MI. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genes (KEGG) enrichment analyses were performed. The STRING database was used to obtain the protein-protein interaction (PPI) network, and Cytoscape was used to identify core targets, transcription factors, and modules. Results: A total of 198 targets were retrieved for the three drugs and 511 targets for T2DM with MI. Finally, 51 related targets, including 31 intersection targets and 20 associated targets, were predicted to interfere with the progression of T2DM and MI on using GLP-1RAs. The STRING database was used to establish a PPI network comprising 46 nodes and 175 edges. The PPI network was analyzed using Cytoscape, and seven core targets were screened: AGT, TGFB1, STAT3, TIMP1, MMP9, MMP1, and MMP2. The transcription factor MAFB regulates all seven core targets. The cluster analysis generated three modules. The GO analysis for 51 targets indicated that the terms were mainly enriched in the extracellular matrix, angiotensin, platelets, and endopeptidase. The results of KEGG analysis revealed that the 51 targets primarily participated in the renin-angiotensin system, complement and coagulation cascades, hypertrophic cardiomyopathy, and AGE-RAGE signaling pathway in diabetic complications. Conclusion: GLP-1RAs exert multi-dimensional effects on reducing the occurrence of MI in T2DM patients by interfering with targets, biological processes, and cellular signaling pathways related to atheromatous plaque, myocardial remodeling, and thrombosis.
Collapse
Affiliation(s)
- Guorong Deng
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiajia Ren
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ruohan Li
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Minjie Li
- Department of Cardiology, The Second Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Xi’an, China
| | - Xuting Jin
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jiamei Li
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jueheng Liu
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Ya Gao
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jingjing Zhang
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaochuang Wang
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Gang Wang
- Department of Critical Care Medicine, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China,*Correspondence: Gang Wang,
| |
Collapse
|
181
|
Xin JJ, Gao JH, Liu Q, Zhao YX, Zhou C, Yu XC. Involvement of Interleukin-1 β/Insulin-Like Growth Factor 1 in Ameliorating Effects of Electroacupuncture on Myocardial Fibrosis Induced by Essential Hypertension. Chin J Integr Med 2023; 29:162-169. [PMID: 35840854 DOI: 10.1007/s11655-022-2897-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2022] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To investigate the effect of electroacupuncture (EA) at Neiguan (PC 6) on myocardial fibrosis in spontaneously hypertensive rats (SHRs), and to explore the contribution of interleukin-1 β (IL-1 β), insulin-like growth factor 1 (IGF-1), and transforming growth factor β 1 (TGF- β 1) to the effects. METHODS Nine 12-weeks-old Wistar Kyoto (WKY) male rats were employed as the normal group. Twenty-seven SHRs were equally randomized into SHR, SHR+EA, and SHR + sham groups. EA was applied at bilateral PC 6 once a day 30 min per day in 8 consecutive weeks. After 8-weeks EA treatment at PC 6, histopathologic changes of collagen type I (Col I), collagen type 1 (Col 1) and the levels of IGF-1, 1L-1 β, TGF- β 1, matrix metalloproteinase (MMP)-2 and MMP-9 were examined in myocardial tissure respectively. RESULTS After 8-weeks EA treatment at PC 6, the enhanced myocardial fibrosis in SHRs were characterized by the increased mean fluorescence intensity of Col I and Col 1 in myocardium tissue (P<0.01). All these abnormal alterations above in SHR + EA group was significantly lower compared with the SHR group (P<0.01). Meanwhile, the increased levels of IL-1 β, IGF-1, TGF-β 1 in serum or myocardial tissue of SHRs, diminished MMP 9 mRNA expression in SHRs were also markedly inhibited after 8 weeks of EA treatment (P<0.05 or P<0.01). Furthermore, the contents of IL-1 β, IGF-1, TGF-β 1 in myocardial tissue were positively correlated with the systolic blood pressure and hydroxyproline respectively (P<0.01). CONCLUSION EA at bilateral PC 6 could ameliorate cardiac fibrosis in SHRs, which might be mediated by regulation of 1L-1 β/IGF-1-TGF- β 1-MMP9 pathway.
Collapse
Affiliation(s)
- Juan-Juan Xin
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jun-Hong Gao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Qun Liu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yu-Xue Zhao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chen Zhou
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiao-Chun Yu
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
182
|
Liu Z, Liu X, Liu L, Wang Y, Zheng J, Li L, Li S, Zhang H, Ni J, Ma C, Gao X, Bian X, Fan G. SUMO1 regulates post-infarct cardiac repair based on cellular heterogeneity. J Pharm Anal 2023; 13:170-186. [PMID: 36908856 PMCID: PMC9999303 DOI: 10.1016/j.jpha.2022.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 11/19/2022] [Accepted: 11/27/2022] [Indexed: 12/11/2022] Open
Abstract
Small ubiquitin-related modifier (SUMOylation) is a dynamic post-translational modification that maintains cardiac function and can protect against a hypertrophic response to cardiac pressure overload. However, the function of SUMOylation after myocardial infarction (MI) and the molecular details of heart cell responses to SUMO1 deficiency have not been determined. In this study, we demonstrated that SUMO1 protein was inconsistently abundant in different cell types and heart regions after MI. However, SUMO1 knockout significantly exacerbated systolic dysfunction and infarct size after myocardial injury. Single-nucleus RNA sequencing revealed the differential role of SUMO1 in regulating heart cells. Among cardiomyocytes, SUMO1 deletion increased the Nppa + Nppb + Ankrd1 + cardiomyocyte subcluster proportion after MI. In addition, the conversion of fibroblasts to myofibroblasts subclusters was inhibited in SUMO1 knockout mice. Importantly, SUMO1 loss promoted proliferation of endothelial cell subsets with the ability to reconstitute neovascularization and expressed angiogenesis-related genes. Computational analysis of ligand/receptor interactions suggested putative pathways that mediate cardiomyocytes to endothelial cell communication in the myocardium. Mice preinjected with cardiomyocyte-specific AAV-SUMO1, but not the endothelial cell-specific form, and exhibited ameliorated cardiac remodeling following MI. Collectively, our results identified the role of SUMO1 in cardiomyocytes, fibroblasts, and endothelial cells after MI. These findings provide new insights into SUMO1 involvement in the pathogenesis of MI and reveal novel therapeutic targets.
Collapse
Affiliation(s)
- Zhihao Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin, 300450, China
| | - Li Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Ying Wang
- Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin, 300450, China
| | - Jie Zheng
- Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin, 300450, China
| | - Lan Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Sheng Li
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China
| | - Han Zhang
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Jingyu Ni
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Chuanrui Ma
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China
| | - Xiumei Gao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Xiyun Bian
- Tianjin Key Laboratory of Epigenetics for Organ Development in Preterm Infants, The Fifth Central Hospital of Tianjin, Tianjin, 300450, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300193, China.,State Key Laboratory of Component-Based Chinese Medicine, Tianjin, 301617, China.,Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| |
Collapse
|
183
|
Ribeiro ASF, Zerolo BE, López-Espuela F, Sánchez R, Fernandes VS. Cardiac System during the Aging Process. Aging Dis 2023:AD.2023.0115. [PMID: 37163425 PMCID: PMC10389818 DOI: 10.14336/ad.2023.0115] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/15/2023] [Indexed: 05/12/2023] Open
Abstract
The aging process is accompanied by a continuous decline of the cardiac system, disrupting the homeostatic regulation of cells, organs, and systems. Aging increases the prevalence of cardiovascular diseases, thus heart failure and mortality. Understanding the cardiac aging process is of pivotal importance once it allows us to design strategies to prevent age-related cardiac events and increasing the quality of live in the elderly. In this review we provide an overview of the cardiac aging process focus on the following topics: cardiac structural and functional modifications; cellular mechanisms of cardiac dysfunction in the aging; genetics and epigenetics in the development of cardiac diseases; and aging heart and response to the exercise.
Collapse
Affiliation(s)
| | - Blanca Egea Zerolo
- Escuela de Enfermería y Fisioterapia San Juan de Dios. Universidad Pontificia Comillas, Madrid, Spain
| | - Fidel López-Espuela
- Metabolic Bone Diseases Research Group, Nursing and Occupational Therapy College, University of Extremadura, Caceres, Spain
| | - Raúl Sánchez
- Unidad de Cardiopatías Congénitas, Hospital Universitario La Paz, Madrid, Spain
| | - Vítor S Fernandes
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
184
|
Zhang M, Lu P, Zhao F, Sun X, Ma W, Tang J, Zhang C, Ji H, Wang X. Uncovering the molecular mechanisms of Curcumae Rhizoma against myocardial fibrosis using network pharmacology and experimental validation. JOURNAL OF ETHNOPHARMACOLOGY 2023; 300:115751. [PMID: 36162550 DOI: 10.1016/j.jep.2022.115751] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Myocardial fibrosis leads to cardiac remodeling and dysfunction. Curcumae Rhizoma has been utilized in clinical trials to treat a variety of cardiovascular illnesses, although its role in myocardial fibrosis is unknown. AIM OF THE STUDY The purpose of current study was to explore the potential mechanism action and anti-myocardial fibrosis effects of treatment with Curcumae Rhizoma. MATERIALS AND METHODS The chemical components in the aqueous extract from Curcumae Rhizoma were identified using GC-MS analysis. A prediction network describing the relationship between Curcumae Rhizoma and MF was established based on information collected from multiple databases. Functional enrichment analysis was performed to investigate the specific functions and pathways involved in the candidate Curcumae Rhizoma targets acting on MF, which were further validated by vivo experiments. RESULTS There were 444 targets obtained from the 39 active ingredients in Curcumae Rhizoma, and 5691 disease targets related to MF were identified. Then, 41 key targets were determined with the PPI interaction network, which was structured from 324 overlapping gene targets. GO and KEGG analyses revealed that the p38 MAPK/NF-κB and TGF-β1/Smad2/3 signaling pathways might play crucial roles in the therapeutic mechanism of MF. According to the results of molecular docking, the binding activity between core components and targets was marvelous (affinity < -6 kcal/mol). Take it a step further, the experimental validation data affirmed that Curcumae Rhizoma substantially decreased myocardial fibrosis and recovered cardiac function in the ISO-induced rats. The associated proteins expression data implied that the p38 MAPK/NF-κB and TGF-β1/Smad2/3 pathways might be vital in the anti-fibrosis effect of Curcumae Rhizoma. CONCLUSION The findings suggested that Curcumae Rhizoma diminished myocardial fibrosis by suppressing fibrosis multiplication and collagen deposition through inhibiting p38 MAPK/NF-κB and TGF-β1/Smad2/3 pathways, which might be a promising therapeutic medicament for alleviating myocardial fibrosis.
Collapse
Affiliation(s)
- Meng Zhang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Pengyu Lu
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Fusen Zhao
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Xuemei Sun
- Changzhou Hospital of Traditional Chinese Medicine, Changzhou, 213000, China
| | - Wenqi Ma
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Jiahui Tang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Chengbo Zhang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China
| | - Haigang Ji
- Changzhou Hospital of Traditional Chinese Medicine, Changzhou, 213000, China.
| | - Xindong Wang
- The Third Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210028, China; Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, China.
| |
Collapse
|
185
|
Song H, Li Y, Yu R, Meng X, Bi Y. Change in left ventricular diastolic function after pioglitazone treatment in patients with type 2 diabetes mellitus: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2023; 102:e32613. [PMID: 36607862 PMCID: PMC9829294 DOI: 10.1097/md.0000000000032613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Pioglitazone is currently used as an anti-diabetic agent and can reduce cardiovascular events in in patients with type 2 diabetes mellitus (T2DM). Left ventricular diastolic dysfunction has been recognized as an early manifestation of myocardial dysfunction in T2DM patients. This systematic review and meta-analysis aimed to investigate changes in the left ventricular diastolic function after the treatment of pioglitazone. METHODS A systematic literature search of PubMed, Embase, and the Cochrane Library until May 2021 with keywords pioglitazone and left ventricular diastolic function was performed in accordance with the meta-analysis of observational studies in epidemiology guidelines and preferred reporting items for systematic reviews and meta-analyses statement. Three reviewers independently selected the studies and extracted data. Quality assessment of the included studies was undergone. A fixed effects model was used to calculate overall effect sizes. Subgroup analyses were subsequently performed. A fixed effects model was used to calculate the overall effect size. Subgroup analyses were then performed. RESULTS Seven studies with 233 patients were investigated. We found pioglitazone significantly improved hemoglobin A1c (%) in patients with T2DM and left ventricular diastolic function had an improvement tendency (weighted mean difference [WMD], 0.03; 95% confidence interval [CI], 0.01-0.05, P < .01) despite moderate heterogeneity (I2 = 66%). Subsequent subgroup analysis indicated that left ventricular diastolic function were significantly improved (WMD, 0.20; 95% CI, 0.12-0.29, P < .001) in T2DM patients whose average age < 55 after receiving pioglitazone treatment. However, in T2DM patients with mean age ≥ 55 years, there was no significant improvement of left ventricular diastolic function (WMD, 0.02; 95% CI, 0-0.04, P = .04). CONCLUSION Pioglitazone treatment significantly improved left ventricular diastolic function in type 2 diabetic patients with a mean age of < 55 years, but did not improve left ventricular diastolic function in patients with a mean age of ≥ 55 years.
Collapse
Affiliation(s)
- Han Song
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yunye Li
- Department of Pharmacy, Qingdao Jiaozhou Central Hospital, Qingdao, Shandong, China
| | - Ruiming Yu
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiangbin Meng
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yanwen Bi
- Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
- * Correspondence: Yanwen Bi, Department of Cardiovascular Surgery, Qilu Hospital of Shandong University, No. 107 West Wenhua Road, Jinan, Shandong 250012, China (e-mail: )
| |
Collapse
|
186
|
Ren LL, Li XJ, Duan TT, Li ZH, Yang JZ, Zhang YM, Zou L, Miao H, Zhao YY. Transforming growth factor-β signaling: From tissue fibrosis to therapeutic opportunities. Chem Biol Interact 2023; 369:110289. [PMID: 36455676 DOI: 10.1016/j.cbi.2022.110289] [Citation(s) in RCA: 56] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 11/09/2022] [Accepted: 11/24/2022] [Indexed: 11/30/2022]
Abstract
Fibrosis refers to the excessive deposition of extracellular matrix components in the processes of wound repair or tissue regeneration after tissue damage. Fibrosis occurs in various organs such as lung, heart, liver, and kidney tissues, resulting in the failure of organ structural integrity and its functional impairment. It has long been thought to be relentlessly progressive and irreversible process, but both preclinical models and clinical trials in multiorgans have shown that fibrosis is a highly dynamic process. Transforming growth factor-beta (TGF-β) is a superfamily of related growth factors. Many studies have described that activation of profibrotic TGF-β signaling promotes infiltration and/or proliferation of preexisting fibroblasts, generation of myofibroblasts, extracellular matrix deposition, and inhibition of collagenolysis, which leads to fibrosis in the pathological milieu. This review describes the effect of TGF-β signaling in fibrotic-associate lung, heart, liver, and kidney tissues, followed by a detailed discussion of canonical and non-canonical TGF-β signaling pathway. In addition, this review also discusses therapeutic options by using natural products and chemical agents, for targeting tissue fibrosis via modulating TGF-β signaling to provide a more specific concept-driven therapy strategy for multiorgan fibrosis.
Collapse
Affiliation(s)
- Li-Li Ren
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Xiao-Jun Li
- Department of Nephrology, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13 Shiliugang Street, Guangzhou, 510315, China
| | - Ting-Ting Duan
- Guangdong Nephrotic Drug Engineering Technology Research Center, Institute of Consun Co. for Chinese Medicine in Kidney Diseases, Guangdong Consun Pharmaceutical Group, Guangzhou, 510530, China
| | - Zheng-Hai Li
- Guangdong Nephrotic Drug Engineering Technology Research Center, Institute of Consun Co. for Chinese Medicine in Kidney Diseases, Guangdong Consun Pharmaceutical Group, Guangzhou, 510530, China
| | - Jun-Zheng Yang
- Guangdong Nephrotic Drug Engineering Technology Research Center, Institute of Consun Co. for Chinese Medicine in Kidney Diseases, Guangdong Consun Pharmaceutical Group, Guangzhou, 510530, China
| | - Ya-Mei Zhang
- Key Disciplines of Clinical Pharmacy, Clinical Genetics Laboratory, Affiliated Hospital & Clinical Medical College of Chengdu University, No. 82 the Second Section of North 2nd Ring Road, Chengdu, Sichuan, 610081, China
| | - Liang Zou
- School of Food and Bioengineering, Chengdu University, No. 2025 Chengluo Avenue, Chengdu, Sichuan, 610106, China
| | - Hua Miao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China.
| | - Ying-Yong Zhao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China; Key Disciplines of Clinical Pharmacy, Clinical Genetics Laboratory, Affiliated Hospital & Clinical Medical College of Chengdu University, No. 82 the Second Section of North 2nd Ring Road, Chengdu, Sichuan, 610081, China.
| |
Collapse
|
187
|
Wang F, Zhao H, Yin L, Zhang W, Tang Y, Wang X, Huang C. The paired-related homeobox protein 1 promotes cardiac fibrosis via the Twist1-Prrx1-tenascin-C loop. Cell Biol Int 2023; 47:167-177. [PMID: 36273425 DOI: 10.1002/cbin.11944] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/13/2022] [Indexed: 12/31/2022]
Abstract
Cardiac fibrosis is a common pathology in the advanced stage of cardiovascular diseases, which leads to cardiac systolic and diastolic dysfunction. It is important to prevent cardiac fibrosis during myocardial injury. The transcription factor Prrx1 is involved in cancer-associated fibrosis and other organ fibrosis. However, the role and mechanism of Prrx1 in cardiac fibrosis deserves further exploration. We identified that overexpressed Prrx1 promoted the proliferation and migration of cardiac fibroblasts, and transform cardiac fibroblasts to myofibroblasts in vitro. We demonstrated that the expression of Prrx1 is upregulated in TGF-β1-treated fibroblasts. And silencing Prrx1 could attenuate cardiac fibrosis induced by TGF-β1 in vitro. In addition, a Twist1-paired-related homeobox 1 (Prrx1)-tenascin-C (TNC) positive feedback loop (PFL) combined with Twist1, Prrx1, and TNC activated fibroblasts, which was the mechanism the Prrx1 in cardiac fibrosis. In conclusion, our findings showed that the deficiency of Prrx1 attenuated cardiac fibrosis in vitro and reveal a novel Twist1-Prrx1-TNC PFL in the regulation of cardiac fibrosis.
Collapse
Affiliation(s)
- Fengyuan Wang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Hongyi Zhao
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Lin Yin
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Wei Zhang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Yanhong Tang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Xi Wang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Hubei Key Laboratory of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Wuhan, China
| |
Collapse
|
188
|
Liu Z, Wang Y, Liu F, Zhu D, Chen Y, Yim WY, Hu K, Rao Z, Pan X, Li F, Dong N. Long noncoding TSI attenuates aortic valve calcification by suppressing TGF-β1-induced osteoblastic differentiation of valve interstitial cells. Metabolism 2023; 138:155337. [PMID: 36273649 DOI: 10.1016/j.metabol.2022.155337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION Calcific aortic valve disease (CAVD) is an active and cellular-driven fibrocalcific process characterised by differentiation of valve interstitial cells (VICs) towards an osteogenic-like phenotype. A recently identified lncRNA, lncTSI, has been reported to inhibit fibrogenesis through transforming growth factor (TGF)-β/Smad3 pathway. Here, the present study aimed to investigate the role of lncTSI in CAVD. METHODS The effect of TGF-β1 on lncTSI of VICs was measured. TGF-β1, RUNX2 and collagen I expression between calcified aortic valve tissue and normal samples by immunohistochemistry and western blotting. Human VICs were cultured and treated with TGF-β1. SiRNA and pcDNA3.1-lncTSI plasmid transfection were used to silence and overexpress lncTSI in VICs for 48 h, Smads phosphorylation, RUNX2 and collagen I expression were then verified by western blotting. In ApoE-/- mice fed with 0.25 % high-cholesterol diet, AAV2-lncTSI were injected intravenously to observe their effect on the formation of aortic valve calcification. RESULTS lncTSI was highly expressed in VICs treated with TGF-β1. lncTSI was transcriptionally regulated by Smad3 and reversely inhibited TGF-β1-induced Smad3 phosphorylation and downregulated profibrotic gene expression. Silencing lncTSI increased TGF-β1-induced Smad3 phosphorylation, and subsequently, upregulated RUNX2 and collagen I expressions in VICs. While overexpression of lncTSI reversed the production of RUNX2 and collagen I in VICs. In a mouse CAVD model of 24 week 0.25 % high-cholesterol diet feeding, overexpression of lncTSI significantly reduced calcium deposition, RUNX2, pSmad3, and collagen I expression in aortic valve leaflets, with less aortic valve stenosis. CONCLUSIONS The novel findings of present study suggested that lncTSI alleviated aortic valve calcification through negative regulation of the TGF-β/Smad3 pathway. The results may help elucidate new diagnostic and therapeutic targets to prevent CAVD progression.
Collapse
Affiliation(s)
- Zongtao Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Fayuan Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Da Zhu
- Structural Heart Center, Fuwai Yunnan Cardiovascular Hospital, Kunming Medical University, 528 Shahebei Rd, 65000 Kunming, China
| | - Yuqi Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wei Yen Yim
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ke Hu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhenqi Rao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiangbin Pan
- Structural Heart Center, Fuwai Yunnan Cardiovascular Hospital, Kunming Medical University, 528 Shahebei Rd, 65000 Kunming, China.
| | - Fei Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
189
|
Cheng Y, Wang Y, Yin R, Xu Y, Zhang L, Zhang Y, Yang L, Zhao D. Central role of cardiac fibroblasts in myocardial fibrosis of diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2023; 14:1162754. [PMID: 37065745 PMCID: PMC10102655 DOI: 10.3389/fendo.2023.1162754] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023] Open
Abstract
Diabetic cardiomyopathy (DCM), a main cardiovascular complication of diabetes, can eventually develop into heart failure and affect the prognosis of patients. Myocardial fibrosis is the main factor causing ventricular wall stiffness and heart failure in DCM. Early control of myocardial fibrosis in DCM is of great significance to prevent or postpone the progression of DCM to heart failure. A growing body of evidence suggests that cardiomyocytes, immunocytes, and endothelial cells involve fibrogenic actions, however, cardiac fibroblasts, the main participants in collagen production, are situated in the most central position in cardiac fibrosis. In this review, we systematically elaborate the source and physiological role of myocardial fibroblasts in the context of DCM, and we also discuss the potential action and mechanism of cardiac fibroblasts in promoting fibrosis, so as to provide guidance for formulating strategies for prevention and treatment of cardiac fibrosis in DCM.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dong Zhao
- *Correspondence: Longyan Yang, ; Dong Zhao,
| |
Collapse
|
190
|
Gao Z, Bao J, Hu Y, Tu J, Ye L, Wang L. Sodium-glucose Cotransporter 2 Inhibitors and Pathological Myocardial Hypertrophy. Curr Drug Targets 2023; 24:1009-1022. [PMID: 37691190 PMCID: PMC10879742 DOI: 10.2174/1389450124666230907115831] [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: 05/02/2023] [Revised: 08/18/2023] [Accepted: 08/23/2023] [Indexed: 09/12/2023]
Abstract
Sodium-glucose cotransporter 2 (SGLT2) inhibitors are a new type of oral hypoglycemic drugs that exert a hypoglycemic effect by blocking the reabsorption of glucose in the proximal renal tubules, thus promoting the excretion of glucose from urine. Their hypoglycemic effect is not dependent on insulin. Increasing data shows that SGLT2 inhibitors improve cardiovascular outcomes in patients with type 2 diabetes. Previous studies have demonstrated that SGLT2 inhibitors can reduce pathological myocardial hypertrophy with or without diabetes, but the exact mechanism remains to be elucidated. To clarify the relationship between SGLT2 inhibitors and pathological myocardial hypertrophy, with a view to providing a reference for the future treatment thereof, this study reviewed the possible mechanisms of SGLT2 inhibitors in attenuating pathological myocardial hypertrophy. We focused specifically on the mechanisms in terms of inflammation, oxidative stress, myocardial fibrosis, mitochondrial function, epicardial lipids, endothelial function, insulin resistance, cardiac hydrogen and sodium exchange, and autophagy.
Collapse
Affiliation(s)
- Zhicheng Gao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiaqi Bao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yilan Hu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Junjie Tu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Lifang Ye
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Lihong Wang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
- Heart Center, Department of Cardiovascular Medicine, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
191
|
Zhu X, Wang C, Duan X, Liang B, Genbo Xu E, Huang Z. Micro- and nanoplastics: A new cardiovascular risk factor? ENVIRONMENT INTERNATIONAL 2023; 171:107662. [PMID: 36473237 DOI: 10.1016/j.envint.2022.107662] [Citation(s) in RCA: 76] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Exposure to micro- and nanoplastics (MNPs) is inevitable due to their omnipresence in the environment. A growing body of studies has advanced our understanding of the potential toxicity of MNPs but knowledge gaps still exist regarding the adverse effects of MNPs on the cardiovascular system and underlying mechanisms, particularly in humans. Here, we reviewed up-to-date data published in the past 10 years on MNP-driven cardiovascular toxicity and mechanisms. Forty-six articles concerning ADME (absorption, distribution, and aggregation behaviors) and toxicity of MNPs in the circulatory system of animals and human cells were analyzed and summarized. The results showed that MNPs affected cardiac functions and caused toxicity on (micro)vascular sites. Direct cardiac toxicity of MNPs included abnormal heart rate, cardiac function impairment, pericardial edema, and myocardial fibrosis. On (micro)vascular sites, MNPs induced hemolysis, thrombosis, blood coagulation, and vascular endothelial damage. The main mechanisms included oxidative stress, inflammation, apoptosis, pyroptosis, and interaction between MNPs and multiple cellular components. Cardiovascular toxicity was determined by the properties (type, size, surface, and structure) of MNPs, exposure dose and duration, protein presence, the life stage, sex, and species of the tested organisms, as well as the interaction with other environmental contamination. The limited quantitative information on MNPs' ADME and the lack of guidelines for MNP cardiotoxicity testing makes risk assessment on cardiac health impossible. Furthermore, the future directions of cardiovascular research on MNPs are recommended to enable more realistic health risk assessment.
Collapse
Affiliation(s)
- Xiaoqi Zhu
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Chuanxuan Wang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyu Duan
- Department of Biology, University of Southern Denmark, Odense 5230, Denmark
| | - Boxuan Liang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Elvis Genbo Xu
- Department of Biology, University of Southern Denmark, Odense 5230, Denmark.
| | - Zhenlie Huang
- NMPA Key Laboratory for Safety Evaluation of Cosmetics, Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
192
|
Endothelial-cell-mediated mechanism of coronary microvascular dysfunction leading to heart failure with preserved ejection fraction. Heart Fail Rev 2023; 28:169-178. [PMID: 35266091 PMCID: PMC9902427 DOI: 10.1007/s10741-022-10224-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/23/2022] [Indexed: 02/07/2023]
Abstract
Although the prevalence of heart failure with preserved ejection fraction (HFpEF) is growing worldwide, its complex pathophysiology has yet to be fully elucidated, and multiple hypotheses have all failed to produce a viable target for therapeutic action or provide effective treatment. Cardiac remodeling has long been considered an important mechanism of HFpEF. Strong evidence has been reported over the past years that coronary microvascular dysfunction (CMD), manifesting as structural and functional abnormalities of coronary microvasculature, also contributes to the evolution of HFpEF. However, the mechanisms of CMD are still not well understood and need to be studied further. Coronary microvascular endothelial cells (CMECs) are one of the most abundant cell types in the heart by number and active players in cardiac physiology and pathology. CMECs are not only important cellular mediators of cardiac vascularization but also play an important role in disease pathophysiology by participating in the inception and progression of cardiac remodeling. CMECs are also actively involved in the pathogenesis of CMD. Numerous studies have confirmed that CMD is closely related to cardiac remodeling. ECs may serve a critical function in mediating the connection between CMD and HFpEF. It follows that CMECs participate in the mechanism of CMD leading to HFpEF. In this review article, we focus on the role of CMD in the pathogenesis of HFpEF resulting from cardiac remodeling and highlight the subsequent complexity of the EC-mediated correlation between CMD and HFpEF.
Collapse
|
193
|
Zou S, Khoo BL. Subtyping based on immune cell fractions reveal heterogeneity of cardiac fibrosis in end-stage heart failure. Front Immunol 2023; 14:1053793. [PMID: 36875078 PMCID: PMC9975711 DOI: 10.3389/fimmu.2023.1053793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023] Open
Abstract
Background A central issue hindering the development of effective anti-fibrosis drugs for heart failure is the unclear interrelationship between fibrosis and the immune cells. This study aims at providing precise subtyping of heart failure based on immune cell fractions, elaborating their differences in fibrotic mechanisms, and proposing a biomarker panel for evaluating intrinsic features of patients' physiological statuses through subtype classification, thereby promoting the precision medicine for cardiac fibrosis. Methods We inferred immune cell type abundance of the ventricular samples by a computational method (CIBERSORTx) based on ventricular tissue samples from 103 patients with heart failure, and applied K-means clustering to divide patients into two subtypes based on their immune cell type abundance. We also designed a novel analytic strategy: Large-Scale Functional Score and Association Analysis (LAFSAA), to study fibrotic mechanisms in the two subtypes. Results Two subtypes of immune cell fractions: pro-inflammatory and pro-remodeling subtypes, were identified. LAFSAA identified 11 subtype-specific pro-fibrotic functional gene sets as the basis for personalised targeted treatments. Based on feature selection, a 30-gene biomarker panel (ImmunCard30) established for diagnosing patient subtypes achieved high classification performance, with the area under the receiver operator characteristic curve corresponding to 0.954 and 0.803 for the discovery and validation sets, respectively. Conclusion Patients with the two subtypes of cardiac immune cell fractions were likely having different fibrotic mechanisms. Patients' subtypes can be predicted based on the ImmunCard30 biomarker panel. We envision that our unique stratification strategy revealed in this study will unravel advance diagnostic techniques for personalised anti-fibrotic therapy.
Collapse
Affiliation(s)
- Shangjie Zou
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, Hong Kong SAR, China
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Hong Kong Center for Cerebro-Cardiovascular Health Engineering (COCHE), Hong Kong, Hong Kong SAR, China.,Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong-Shenzhen Futian Research Institute, Shenzhen, China
| |
Collapse
|
194
|
Mostert D, Groenen B, Klouda L, Passier R, Goumans MJ, Kurniawan NA, Bouten CVC. Human pluripotent stem cell-derived cardiomyocytes align under cyclic strain when guided by cardiac fibroblasts. APL Bioeng 2022; 6:046108. [PMID: 36567768 PMCID: PMC9771596 DOI: 10.1063/5.0108914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
The myocardium is a mechanically active tissue typified by anisotropy of the resident cells [cardiomyocytes (CMs) and cardiac fibroblasts (cFBs)] and the extracellular matrix (ECM). Upon ischemic injury, the anisotropic tissue is replaced by disorganized scar tissue, resulting in loss of coordinated contraction. Efforts to re-establish tissue anisotropy in the injured myocardium are hampered by a lack of understanding of how CM and/or cFB structural organization is affected by the two major physical cues inherent in the myocardium: ECM organization and cyclic mechanical strain. Herein, we investigate the singular and combined effect of ECM (dis)organization and cyclic strain in a two-dimensional human in vitro co-culture model of the myocardial microenvironment. We show that (an)isotropic ECM protein patterning can guide the orientation of CMs and cFBs, both in mono- and co-culture. Subsequent application of uniaxial cyclic strain-mimicking the local anisotropic deformation of beating myocardium-causes no effect when applied parallel to the anisotropic ECM. However, when cultured on isotropic substrates, cFBs, but not CMs, orient away from the direction of cyclic uniaxial strain (strain avoidance). In contrast, CMs show strain avoidance via active remodeling of their sarcomeres only when co-cultured with at least 30% cFBs. Paracrine signaling or N-cadherin-mediated communication between CMs and cFBs was no contributing factor. Our findings suggest that the mechanoresponsive cFBs provide structural guidance for CM orientation and elongation. Our study, therefore, highlights a synergistic mechanobiological interplay between CMs and cFBs in shaping tissue organization, which is of relevance for regenerating functionally organized myocardium.
Collapse
Affiliation(s)
| | - Bart Groenen
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Leda Klouda
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | | | - Marie-Jose Goumans
- Department of Cell and Chemical Biology and Center for Biomedical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | | |
Collapse
|
195
|
Tu Y, Li Q, Zhou Y, Ye Z, Wu C, Xie E, Li Y, Li P, Wu Y, Guo Z, Yu C, Zheng J, Gao Y. Empagliflozin inhibits coronary microvascular dysfunction and reduces cardiac pericyte loss in db/db mice. Front Cardiovasc Med 2022; 9:995216. [PMID: 36588571 PMCID: PMC9800791 DOI: 10.3389/fcvm.2022.995216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Background Coronary microvascular dysfunction (CMD) is a pathophysiological feature of diabetic heart disease. However, whether sodium-glucose cotransporter 2 (SGLT2) inhibitors protect the cardiovascular system by alleviating CMD is not known. Objective We observed the protective effects of empagliflozin (EMPA) on diabetic CMD. Materials and methods The mice were randomly divided into a db/db group and a db/db + EMPA group, and db/m mice served as controls. At 8 weeks of age, the db/db + EMPA group was given empagliflozin 10 mg/(kg⋅d) by gavage for 8 weeks. Body weight, fasting blood glucose and blood pressure were dynamically observed. Cardiac systolic and diastolic function and coronary flow reserve (CFR) were detected using echocardiography. The coronary microvascular structure and distribution of cardiac pericytes were observed using immunofluorescence staining. Picrosirius red staining was performed to evaluate cardiac fibrosis. Results Empagliflozin lowered the increased fasting blood glucose levels of the db/db group. The left ventricular ejection fraction, left ventricular fractional shortening, E/A ratio and E/e' ratio were not significantly different between the three groups. CFR was decreased in the db/db group, but EMPA significantly improved CFR. In contrast to the sparse and abnormal expansion of coronary microvessels observed in the db/db group, the number of coronary microvessels was increased, and the capillary diameter was decreased in the db/db + EMPA group. The number and microvascular coverage of cardiac pericytes were reduced in the db/db mice but were improved by EMPA. The cardiac fibrosis was increased in db/db group and may alleviate by EMPA. Conclusion Empagliflozin inhibited CMD and reduced cardiac pericyte loss in diabetic mice.
Collapse
Affiliation(s)
- Yimin Tu
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qing Li
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yuanchen Zhou
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Zixiang Ye
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Chao Wu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Enmin Xie
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yike Li
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Peizhao Li
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Yaxin Wu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Ziyu Guo
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Changan Yu
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Jingang Zheng
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| | - Yanxiang Gao
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
- Department of Cardiology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
196
|
Proteomic Insights into Cardiac Fibrosis: From Pathophysiological Mechanisms to Therapeutic Opportunities. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248784. [PMID: 36557919 PMCID: PMC9781843 DOI: 10.3390/molecules27248784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Cardiac fibrosis is a common pathophysiologic process in nearly all forms of heart disease which refers to excessive deposition of extracellular matrix proteins by cardiac fibroblasts. Activated fibroblasts are the central cellular effectors in cardiac fibrosis, and fibrotic remodelling can cause several cardiac dysfunctions either by reducing the ejection fraction due to a stiffened myocardial matrix, or by impairing electric conductance. Recently, there is a rising focus on the proteomic studies of cardiac fibrosis for pathogenesis elucidation and potential biomarker mining. This paper summarizes the current knowledge of molecular mechanisms underlying cardiac fibrosis, discusses the potential of imaging and circulating biomarkers available to recognize different phenotypes of this lesion, reviews the currently available and potential future therapies that allow individualized management in reversing progressive fibrosis, as well as the recent progress on proteomic studies of cardiac fibrosis. Proteomic approaches using clinical specimens and animal models can provide the ability to track pathological changes and new insights into the mechanisms underlining cardiac fibrosis. Furthermore, spatial and cell-type resolved quantitative proteomic analysis may also serve as a minimally invasive method for diagnosing cardiac fibrosis and allowing for the initiation of prophylactic treatment.
Collapse
|
197
|
A Study on the Protective Effect of sRAGE-MSCs in a Rodent Reperfusion Model of Myocardial Infarction. Int J Mol Sci 2022; 23:ijms232415630. [PMID: 36555270 PMCID: PMC9779272 DOI: 10.3390/ijms232415630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Acute myocardial infarction (AMI) is one of the major leading causes of death in humans globally. Recently, increased levels of recruited macrophages and AGE-albumin were observed in the hearts of humans and animals with acute myocardial infarction. Thus, the purposes of this study were to investigate whether the elevated levels of AGE-albumin from activated macrophage cells are implicated in ischemia-induced cardiomyocyte death and to develop therapeutic strategies for AMI based on its underlying molecular mechanisms with respect to AGEs. The present study demonstrated that activated macrophages and AGE-albumin were observed in heart tissues obtained from humans and rats with AMI incidences. In the cellular model of AMI, it was found that increased expression of AGE-albumin was shown to be co-localized with macrophages, and the presence of AGE-albumin led to increased expression of RAGE through the mitogen-activated protein kinase pathway. After revealing cardiomyocyte apoptosis induced by toxicity of the AGE-RAGE system, sRAGE-secreting MSCs were generated using the CRISPR/Cas9 platform to investigate the therapeutic effects of sRAGE-MSCs in an AMI rat model. Gene-edited sRAGE-MSCs showed greater therapeutic effects against AMI pathogenesis in rat models compared to mock MSCs, and promising results of the functional improvement of stem cells could result in significant improvements in the clinical management of cardiovascular diseases.
Collapse
|
198
|
Loss of KDM5B ameliorates pathological cardiac fibrosis and dysfunction by epigenetically enhancing ATF3 expression. Exp Mol Med 2022; 54:2175-2187. [PMID: 36481938 PMCID: PMC9794816 DOI: 10.1038/s12276-022-00904-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/26/2022] [Accepted: 10/24/2022] [Indexed: 12/13/2022] Open
Abstract
Excessive cardiac fibrosis is central to adverse cardiac remodeling and dysfunction leading to heart failure in many cardiac diseases. Histone methylation plays a crucial role in various pathophysiological events. However, the role of histone methylation modification enzymes in pathological cardiac fibrosis needs to be fully elucidated. Here, we identified lysine demethylase 5B (KDM5B), a histone H3K4me2/me3 demethylase, as a key epigenetic mediator of pathological cardiac fibrosis. KDM5B expression was upregulated in cardiac fibroblasts and myocardial tissues in response to pathological stress. KDM5B deficiency markedly ameliorated cardiac fibrosis, improved cardiac function, and prevented adverse cardiac remodeling following myocardial infarction (MI) or pressure overload. KDM5B knockout or inhibitor treatment constrained the transition of cardiac fibroblasts to profibrogenic myofibroblasts and suppressed fibrotic responses. KDM5B deficiency also facilitated the transformation of cardiac fibroblasts to endothelial-like cells and promoted angiogenesis in response to myocardial injury. Mechanistically, KDM5B bound to the promoter of activating transcription factor 3 (Atf3), an antifibrotic regulator of cardiac fibrosis, and inhibited ATF3 expression by demethylating the activated H3K4me2/3 modification, leading to the enhanced activation of TGF-β signaling and excessive expression of profibrotic genes. Our study indicates that KDM5B drives pathological cardiac fibrosis and represents a candidate target for intervention in cardiac dysfunction and heart failure.
Collapse
|
199
|
Moita MR, Silva MM, Diniz C, Serra M, Hoet RM, Barbas A, Simão D. Transcriptome and proteome profiling of activated cardiac fibroblasts supports target prioritization in cardiac fibrosis. Front Cardiovasc Med 2022; 9:1015473. [PMID: 36531712 PMCID: PMC9751336 DOI: 10.3389/fcvm.2022.1015473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/15/2022] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Activated cardiac fibroblasts (CF) play a central role in cardiac fibrosis, a condition associated with most cardiovascular diseases. Conversion of quiescent into activated CF sustains heart integrity upon injury. However, permanence of CF in active state inflicts deleterious heart function effects. Mechanisms underlying this cell state conversion are still not fully disclosed, contributing to a limited target space and lack of effective anti-fibrotic therapies. MATERIALS AND METHODS To prioritize targets for drug development, we studied CF remodeling upon activation at transcriptomic and proteomic levels, using three different cell sources: primary adult CF (aHCF), primary fetal CF (fHCF), and induced pluripotent stem cells derived CF (hiPSC-CF). RESULTS All cell sources showed a convergent response upon activation, with clear morphological and molecular remodeling associated with cell-cell and cell-matrix interactions. Quantitative proteomic analysis identified known cardiac fibrosis markers, such as FN1, CCN2, and Serpine1, but also revealed targets not previously associated with this condition, including MRC2, IGFBP7, and NT5DC2. CONCLUSION Exploring such targets to modulate CF phenotype represents a valuable opportunity for development of anti-fibrotic therapies. Also, we demonstrate that hiPSC-CF is a suitable cell source for preclinical research, displaying significantly lower basal activation level relative to primary cells, while being able to elicit a convergent response upon stimuli.
Collapse
Affiliation(s)
- Maria Raquel Moita
- iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Oeiras, Portugal
| | - Marta M. Silva
- iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| | - Cláudia Diniz
- iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Oeiras, Portugal
| | - Margarida Serra
- iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Oeiras, Portugal
| | - René M. Hoet
- Department of Pathology, CARIM - School of Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
| | | | - Daniel Simão
- iBET - Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal
| |
Collapse
|
200
|
Chang T, Liu C, Yang H, Lu K, Han Y, Zheng Y, Huang H, Wu Y, Song Y, Yu Q, Shen Z, Jiang T, Zhang Y. Fibrin-based cardiac patch containing neuregulin-1 for heart repair after myocardial infarction. Colloids Surf B Biointerfaces 2022; 220:112936. [DOI: 10.1016/j.colsurfb.2022.112936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/11/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022]
|