1
|
Meng X, Du W, Sun Z. Fine particulate matter‑induced cardiac developmental toxicity (Review). Exp Ther Med 2025; 29:6. [PMID: 39534282 PMCID: PMC11552469 DOI: 10.3892/etm.2024.12756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
Fine particulate matter (PM2.5) has become an important risk factor threatening human health. Epidemiological and toxicological investigations have revealed that PM2.5 not only leads to cardiovascular dysfunction, but it also gives rise to various adverse health effects on the human body, such as cardiovascular and cerebrovascular diseases, cancers, neurodevelopmental disorders, depression and autism. PM2.5 is able to penetrate both respiratory and placental barriers, thereby resulting in negative effects on fetal development. A large body of epidemiological evidences has suggested that gestational exposure to PM2.5 increases the incidence of congenital diseases in offspring, including congenital heart defects. In addition, animal model studies have revealed that gestational exposure to PM2.5 can disrupt normal heart development in offspring, although the potential molecular mechanisms have yet to be fully elucidated. The aim of the present review was to provide a brief overview of what is currently known regarding the molecular mechanisms underlying cardiac developmental toxicity in offspring induced by gestational exposure to PM2.5.
Collapse
Affiliation(s)
- Xiangjiang Meng
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| | - Weiyuan Du
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| | - Zongli Sun
- Department of Cardiovascular Medicine, Changle People's Hospital, Shandong Second Medical University, Weifang, Shandong 262400, P.R. China
| |
Collapse
|
2
|
Zhang Q, Miao M, Cao S, Liu D, Cao Z, Bai X, Yin Y, Jin S, Dong L, Zheng M. PCSK9 promotes vascular neointimal hyperplasia through non-lipid regulation of vascular smooth muscle cell proliferation, migration, and autophagy. Biochem Biophys Res Commun 2025; 742:151081. [PMID: 39632291 DOI: 10.1016/j.bbrc.2024.151081] [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: 09/25/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
We aim to explore the impact of Proprotein convertase subtilisin-kexin type 9 (PCSK9) and its inhibitor evolocumab on neointimal hyperplasia. Wild type and PCSK9 knockout (PCSK9-/-) mice were subjected to ligation of the common carotid artery, with or without subcutaneous injection of evolocumab. Mouse aortic vascular smooth muscle (MOVAS) cells were pretreated with evolocumab or under siRNA-mediated suppression of PCSK9, and then exposed to platelet-derived growth factor type BB(PDGF-BB), a major promoter of MOVAS transformation to a proliferative phenotype. PCSK9 was upregulated in ligated carotid arteries and PDGF-BB-treated MOVAS cells. PCSK9-/- mice showed decreased intimal area and intimal/media area ratio, downregulation of proliferation and autophagy, which was coincidence with wild-type mice treated with evolocumab. In MOVAS cells fortified with evolocumab or silencing of PCSK9, PCNA, Beclin1, p62, LC3 were downregulated, additionally, EdU-positive cells decreased, cell viability reduced, migration ability was weakened, and the number of autophagosomes and autolysosomes decreased after the treatment. We also identified the PI3K/AKT/mTOR signaling molecules as potential PCSK9 targets mediating proliferative effect in MOVAS cells. To sum up, our results suggest that PCSK9 has intrinsic properties to promote proliferation, migration and autophagy in VSMCs independent of its lipid-regulating role. The proliferative effects of PCSK9 may be mediated by the PI3K/AKT/mTOR signaling pathway. These data provide additional evidence for PCSK9i in cardiovascular disease beyond the low-density lipoprotein (LDL)-lowering benefit.
Collapse
MESH Headings
- Animals
- Autophagy
- Cell Proliferation
- Proprotein Convertase 9/metabolism
- Proprotein Convertase 9/genetics
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Cell Movement
- Neointima/metabolism
- Neointima/pathology
- Mice
- Hyperplasia/metabolism
- Hyperplasia/pathology
- Antibodies, Monoclonal, Humanized/pharmacology
- Male
- Mice, Knockout
- Mice, Inbred C57BL
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Becaplermin/metabolism
- Becaplermin/pharmacology
- Signal Transduction
Collapse
Affiliation(s)
- Qian Zhang
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, Hebei, China
| | - Mengdan Miao
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, Hebei, China; Department of Cardiology, Handan First Hospital, Handan, 056000, Hebei, China
| | - Shanhu Cao
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, Hebei, China
| | - Da Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, Hebei, China
| | - Zelong Cao
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, Hebei, China
| | - Xiaoyu Bai
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, Hebei, China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, Hebei, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, 361 zhongshan Road, Shijiazhuang, 050017, China
| | - Lihua Dong
- Department of Biochemistry and Molecular Biology, Hebei Medical University, 050017, Shijiazhuang, China
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, 050031, Hebei, China.
| |
Collapse
|
3
|
Sniezek Carney O, Harris KW, Wohlfarter Y, Lee K, Butschek G, Anzmann AF, Hamacher-Brady A, Keller MA, Vernon HJ. Stem cell models of TAFAZZIN deficiency reveal novel tissue-specific pathologies in Barth syndrome. Hum Mol Genet 2024:ddae152. [PMID: 39535077 DOI: 10.1093/hmg/ddae152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/30/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Barth syndrome (BTHS) is a rare mitochondrial disease caused by pathogenic variants in the gene TAFAZZIN, which leads to abnormal cardiolipin (CL) metabolism on the inner mitochondrial membrane. Although TAFAZZIN is ubiquitously expressed, BTHS involves a complex combination of tissue specific phenotypes including cardiomyopathy, neutropenia, skeletal myopathy, and growth delays, with a relatively minimal neurological burden. To understand both the developmental and functional effects of TAZ-deficiency in different tissues, we generated isogenic TAZ knockout (TAZ-KO) and WT cardiomyocytes (CMs) and neural progenitor cells (NPCs) from CRISPR-edited induced pluripotent stem cells (iPSCs). In TAZ-KO CMs we discovered evidence of dysregulated mitophagy including dysmorphic mitochondria and mitochondrial cristae, differential expression of key autophagy-associated genes, and an inability of TAZ-deficient CMs to properly initiate stress-induced mitophagy. In TAZ-deficient NPCs we identified novel phenotypes including a reduction in CIV abundance and CIV activity in the CIII2&CIV2 intermediate complex. Interestingly, while CL acyl chain manipulation was unable to alter mitophagy defects in TAZ-KO CMs, we found that linoleic acid or oleic acid supplementation was able to partially restore CIV abundance in TAZ-deficient NPCs. Taken together, our results have implications for understanding the tissue-specific pathology of BTHS and potential for tissue-specific therapeutic targeting. Moreover, our results highlight an emerging role for mitophagy in the cardiac pathophysiology of BTHS and reveal a potential neuron-specific bioenergetic phenotype.
Collapse
Affiliation(s)
- Olivia Sniezek Carney
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| | - Kodi W Harris
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| | - Yvonne Wohlfarter
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, Innsbruck 6020, Austria
| | - Kyuna Lee
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| | - Grant Butschek
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Arianna F Anzmann
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| | - Anne Hamacher-Brady
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, United States
| | - Markus A Keller
- Institute of Human Genetics, Medical University of Innsbruck, Peter-Mayr-Str. 1/1.OG, Innsbruck 6020, Austria
| | - Hilary J Vernon
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21205, United States
| |
Collapse
|
4
|
Panico C, Felicetta A, Kunderfranco P, Cremonesi M, Salvarani N, Carullo P, Colombo F, Idini A, Passaretti M, Doro R, Rubino M, Villaschi A, Da Rin G, Peano C, Kallikourdis M, Greco CM, Condorelli G. Single-Cell RNA Sequencing Reveals Metabolic Stress-Dependent Activation of Cardiac Macrophages in a Model of Dyslipidemia-Induced Diastolic Dysfunction. Circulation 2024; 150:1517-1532. [PMID: 38126199 DOI: 10.1161/circulationaha.122.062984] [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: 10/26/2022] [Accepted: 11/17/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Metabolic distress is often associated with heart failure with preserved ejection fraction (HFpEF) and represents a therapeutic challenge. Metabolism-induced systemic inflammation links comorbidities with HFpEF. How metabolic changes affect myocardial inflammation in the context of HFpEF is not known. METHODS We found that ApoE knockout mice fed a Western diet recapitulate many features of HFpEF. Single-cell RNA sequencing was used for expression analysis of CD45+ cardiac cells to evaluate the involvement of inflammation in diastolic dysfunction. We focused bioinformatics analysis on macrophages, obtaining high-resolution identification of subsets of these cells in the heart, enabling us to study the outcomes of metabolic distress on the cardiac macrophage infiltrate and to identify a macrophage-to-cardiomyocyte regulatory axis. To test whether a clinically relevant sodium glucose cotransporter-2 inhibitor could ameliorate the cardiac immune infiltrate profile in our model, mice were randomized to receive the sodium glucose cotransporter-2 inhibitor dapagliflozin or vehicle for 8 weeks. RESULTS ApoE knockout mice fed a Western diet presented with reduced diastolic function, reduced exercise tolerance, and increased pulmonary congestion associated with cardiac lipid overload and reduced polyunsaturated fatty acids. The main immune cell types infiltrating the heart included 4 subpopulations of resident and monocyte-derived macrophages, determining a proinflammatory profile exclusively in ApoE knockout-Western diet mice. Lipid overload had a direct effect on inflammatory gene activation in macrophages, mediated through endoplasmic reticulum stress pathways. Investigation of the macrophage-to-cardiomyocyte regulatory axis revealed the potential effects on cardiomyocytes of multiple inflammatory cytokines secreted by macrophages, affecting pathways such as hypertrophy, fibrosis, and autophagy. Finally, we describe an anti-inflammatory effect of sodium glucose cotransporter-2 inhibition in this model. CONCLUSIONS Using single-cell RNA sequencing in a model of diastolic dysfunction driven by hyperlipidemia, we have determined the effects of metabolic distress on cardiac inflammatory cells, in particular on macrophages, and suggest sodium glucose cotransporter-2 inhibitors as potential therapeutic agents for the targeting of a specific phenotype of HFpEF.
Collapse
Affiliation(s)
- Cristina Panico
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Arianna Felicetta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Paolo Kunderfranco
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Marco Cremonesi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Nicolò Salvarani
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- Institute of Genetics and Biomedical Research, National Research Council of Italy (Milan Unit), Rozzano (MI), Italy (N.S., P.C., C. Peano)
| | - Pierluigi Carullo
- Institute of Genetics and Biomedical Research, National Research Council of Italy (Milan Unit), Rozzano (MI), Italy (N.S., P.C., C. Peano)
| | - Federico Colombo
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Alessandra Idini
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Mauro Passaretti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Riccardo Doro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Marcello Rubino
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Alessandro Villaschi
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Giorgio Da Rin
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Clelia Peano
- Institute of Genetics and Biomedical Research, National Research Council of Italy (Milan Unit), Rozzano (MI), Italy (N.S., P.C., C. Peano)
| | - Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Carolina M Greco
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| | - Gianluigi Condorelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy (C. Panico, A.F., M.C., N.S., A.I., M.P., R,D., A.V., M.K., C.M.G., G.C.)
- IRCCS Humanitas Research Hospital, Rozzano (MI), Italy (C. Panico, A.F., P.K., M.C., F.C., A.I., M.P., R,D., M.R., A.V., G.D.R., M.K., C.M.G., G.C.)
| |
Collapse
|
5
|
Lu C, Liu Y, Ren F, Zhang H, Hou Y, Zhang H, Chen Z, Du X. HO-1: An emerging target in fibrosis. J Cell Physiol 2024:e31465. [PMID: 39420552 DOI: 10.1002/jcp.31465] [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: 05/13/2024] [Revised: 09/08/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Fibrosis, an aberrant reparative response to tissue injury, involves a disruption in the equilibrium between the synthesis and degradation of the extracellular matrix, leading to its excessive accumulation within normal tissues, and culminating in organ dysfunction. Manifesting in the terminal stages of nearly all chronic ailments, fibrosis carries a high mortality rate and poses a significant threat to human health. Heme oxygenase-1 (HO-1) emerges as an endogenous protective agent, mitigating tissue damage through its antioxidant, anti-inflammatory, and antiapoptotic properties. Numerous studies have corroborated HO-1's potential as a therapeutic target in anti-fibrosis treatment. This review delves into the structural and functional attributes, and the upstream and downstream pathways of HO-1. Additionally, the regulatory networks and mechanisms of HO-1 in cells associated with fibrosis are elucidated. The role of HO-1 in various fibrosis-related diseases is also explored. Collectively, this comprehensive information serves as a foundation for future research and augments the viability of HO-1 as a therapeutic target for fibrosis.
Collapse
Affiliation(s)
- Chenxi Lu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Yuan Liu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Feifei Ren
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Haoran Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Yafang Hou
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Hong Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Zhiyong Chen
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Xia Du
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| |
Collapse
|
6
|
Ji Z, Guo J, Zhang R, Zuo W, Xu Y, Qu Y, Tao Z, Li X, Li Y, Yao Y, Ma G. ADAM8 deficiency in macrophages promotes cardiac repair after myocardial infarction via ANXA2-mTOR-autophagy pathway. J Adv Res 2024:S2090-1232(24)00322-9. [PMID: 39097092 DOI: 10.1016/j.jare.2024.07.037] [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: 05/25/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024] Open
Abstract
INTRODUCTION A disintegrin and metalloproteinase 8 (ADAM8), a crucial regulator in macrophages, is closely associated with cardiovascular disease progression. OBJECTIVES This study aimed to explore how ADAM8 regulates macrophage function to inhibit cardiac repair after myocardial infarction (MI). METHODS Macrophage-specific ADAM8 knockout mice (ADAM8flox/flox, Lyz2-Cre, KO) and corresponding control mice (ADAM8flox/flox, Flox) were established using the CRISPR/Cas9 system. Bone marrow transplantation was performed, and macrophage-specific ADAM8-overexpressing adeno-associated virus (AAV6-CD68-Adam8) was produced. Finally, proteomics, RNA sequencing, and co-immunoprecipitation/mass spectrometry (COIP/MS) were used to explore the underlying mechanisms involved. RESULTS ADAM8 was highly expressed in the plasma of patients with acute myocardial infarction (AMI) and in cardiac macrophages derived from AMI mice. ADAM8 KO mice exhibited enhanced angiogenesis, suppressed inflammation, reduced cardiac fibrosis, and improved cardiac function during AMI, which were reversed by overexpressing macrophage-specific ADAM8 and intervention with the clinical anti-angiogenic biologic bevacizumab. Bone marrow transplantation experiments produced ADAM8 KO phenotypes. RNA sequencing showed that autophagy was activated in bone marrow-derived macrophages (BMDMs) with ADAM8 KO, which was confirmed via p-mTOR Ser2448/mTOR, p62, and LC3II/I detection. Autophagy inactivation suppressed angiogenic factor release and promoted inflammation in BMDMs with ADAM8 KO. Mechanistically, ADAM8 could bind to ANXA2 and promote phosphorylation of the ANXA2 Ser26 site. ADAM8 KO impeded ANXA2 phosphorylation, inhibited mTOR Ser2448 site phosphorylation, and activated autophagy, which were demonstrated using the activation or inactivation of ANXA2 phosphorylation. CONCLUSIONS ADAM8 was increased in cardiac macrophages after AMI. The ADAM8-ANXA2-mTOR-autophagy axis in macrophages is responsible for regulating angiogenesis and inflammation following MI. Thus, ADAM8 may be a new target in MI treatment.
Collapse
Affiliation(s)
- Zhenjun Ji
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Jiaqi Guo
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Rui Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Wenjie Zuo
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, Jiangxi, China
| | - Yang Xu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yangyang Qu
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Zaixiao Tao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Xinxin Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yongjun Li
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China.
| |
Collapse
|
7
|
Guo Z. The role of glucagon-like peptide-1/GLP-1R and autophagy in diabetic cardiovascular disease. Pharmacol Rep 2024; 76:754-779. [PMID: 38890260 DOI: 10.1007/s43440-024-00609-1] [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: 11/30/2023] [Revised: 05/25/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024]
Abstract
Diabetes leads to a significantly accelerated incidence of various related macrovascular complications, including peripheral vascular disease and cardiovascular disease (the most common cause of mortality in diabetes), as well as microvascular complications such as kidney disease and retinopathy. Endothelial dysfunction is the main pathogenic event of diabetes-related vascular disease at the earliest stage of vascular injury. Understanding the molecular processes involved in the development of diabetes and its debilitating vascular complications might bring up more effective and specific clinical therapies. Long-acting glucagon-like peptide (GLP)-1 analogs are currently available in treating diabetes with widely established safety and extensively evaluated efficacy. In recent years, autophagy, as a critical lysosome-dependent self-degradative process to maintain homeostasis, has been shown to be involved in the vascular endothelium damage in diabetes. In this review, the GLP-1/GLP-1R system implicated in diabetic endothelial dysfunction and related autophagy mechanism underlying the pathogenesis of diabetic vascular complications are briefly presented. This review also highlights a possible crosstalk between autophagy and the GLP-1/GLP-1R axis in the treatment of diabetic angiopathy.
Collapse
Affiliation(s)
- Zi Guo
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
8
|
Restrepo LJ, Baehrecke EH. Regulation and Functions of Autophagy During Animal Development. J Mol Biol 2024; 436:168473. [PMID: 38311234 PMCID: PMC11260256 DOI: 10.1016/j.jmb.2024.168473] [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: 12/12/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
Autophagy is used to degrade cytoplasmic materials, and is critical to maintain cell and organismal health in diverse animals. Here we discuss the regulation, utilization and impact of autophagy on development, including roles in oogenesis, spermatogenesis and embryogenesis in animals. We also describe how autophagy influences postembryonic development in the context of neuronal and cardiac development, wound healing, and tissue regeneration. We describe recent studies of selective autophagy during development, including mitochondria-selective autophagy and endoplasmic reticulum (ER)-selective autophagy. Studies of developing model systems have also been used to discover novel regulators of autophagy, and we explain how studies of autophagy in these physiologically relevant systems are advancing our understanding of this important catabolic process.
Collapse
Affiliation(s)
- Lucas J Restrepo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA.
| |
Collapse
|
9
|
Maroli G, Schänzer A, Günther S, Garcia-Gonzalez C, Rupp S, Schlierbach H, Chen Y, Graumann J, Wietelmann A, Kim J, Braun T. Inhibition of autophagy prevents cardiac dysfunction at early stages of cardiomyopathy in Bag3-deficient hearts. J Mol Cell Cardiol 2024; 193:53-66. [PMID: 38838815 DOI: 10.1016/j.yjmcc.2024.06.001] [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: 10/17/2023] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
The HSP70 co-chaperone BAG3 targets unfolded proteins to degradation via chaperone assisted selective autophagy (CASA), thereby playing pivotal roles in the proteostasis of adult cardiomyocytes (CMs). However, the complex functions of BAG3 for regulating autophagy in cardiac disease are not completely understood. Here, we demonstrate that conditional inactivation of Bag3 in murine CMs leads to age-dependent dysregulation of autophagy, associated with progressive cardiomyopathy. Surprisingly, Bag3-deficient CMs show increased canonical and non-canonical autophagic flux in the juvenile period when first signs of cardiac dysfunction appear, but reduced autophagy during later stages of the disease. Juvenile Bag3-deficient CMs are characterized by decreased levels of soluble proteins involved in synchronous contraction of the heart, including the gap junction protein Connexin 43 (CX43). Reiterative administration of chloroquine (CQ), an inhibitor of canonical and non-canonical autophagy, but not inactivation of Atg5, restores normal concentrations of soluble cardiac proteins in juvenile Bag3-deficient CMs without an increase of detergent-insoluble proteins, leading to complete recovery of early-stage cardiac dysfunction in Bag3-deficient mice. We conclude that loss of Bag3 in CMs leads to age-dependent differences in autophagy and cardiac dysfunction. Increased non-canonical autophagic flux in the juvenile period removes soluble proteins involved in cardiac contraction, leading to early-stage cardiomyopathy, which is prevented by reiterative CQ treatment.
Collapse
Affiliation(s)
- Giovanni Maroli
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.; Department of Lung Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany..
| | - Anne Schänzer
- Institute of Neuropathology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Bioinformatics and deep sequencing platform, Ludwigstr. 43., 61231 Bad Nauheim, Germany
| | - Claudia Garcia-Gonzalez
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.; Department of Morphology and Cell Biology, University of Oviedo, 33006 Oviedo, Spain
| | - Stefan Rupp
- Department of Pediatric Cardiology and Congenital Heart Disease, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Hannah Schlierbach
- Institute of Neuropathology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Yanpu Chen
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Johannes Graumann
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.; The German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Astrid Wietelmann
- Magnetic Resonance Imaging Group, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Johnny Kim
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany.; The German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main..
| |
Collapse
|
10
|
Ma C, Liu Y, Fu Z. Implications of endoplasmic reticulum stress and autophagy in aging and cardiovascular diseases. Front Pharmacol 2024; 15:1413853. [PMID: 39119608 PMCID: PMC11306071 DOI: 10.3389/fphar.2024.1413853] [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: 04/08/2024] [Accepted: 06/24/2024] [Indexed: 08/10/2024] Open
Abstract
The average lifespan of humans has been increasing, resulting in a rapidly rising percentage of older individuals and high morbidity of aging-associated diseases, especially cardiovascular diseases (CVDs). Diverse intracellular and extracellular factors that interrupt homeostatic functions in the endoplasmic reticulum (ER) induce ER stress. Cells employ a dynamic signaling pathway of unfolded protein response (UPR) to buffer ER stress. Recent studies have demonstrated that ER stress triggers various cellular processes associated with aging and many aging-associated diseases, including CVDs. Autophagy is a conserved process involving lysosomal degradation and recycling of cytoplasmic components, proteins, organelles, and pathogens that invade the cytoplasm. Autophagy is vital for combating the adverse influence of aging on the heart. The present report summarizes recent studies on the mechanism of ER stress and autophagy and their overlap in aging and on CVD pathogenesis in the context of aging. It also discusses possible therapeutic interventions targeting ER stress and autophagy that might delay aging and prevent or treat CVDs.
Collapse
Affiliation(s)
- Chenguang Ma
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Liu
- 32295 Troops of P.L.A, Liaoyang, China
| | - Zhiling Fu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Fang C, Di S, Yu Y, Qi P, Wang X, Jin Y. 6PPD induced cardiac dysfunction in zebrafish associated with mitochondrial damage and inhibition of autophagy processes. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134357. [PMID: 38643584 DOI: 10.1016/j.jhazmat.2024.134357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 04/23/2024]
Abstract
The compound 6PPD is widely acknowledged for its antioxidative properties; however, concerns regarding its impact on aquatic organisms have spurred comprehensive investigations. In our study, we advanced our comprehension by revealing that exposure to 6PPD could induce cardiac dysfunction, myocardial injury and DNA damage in adult zebrafish. Furthermore, our exploration unveiled that the exposure of cardiomyocytes to 6PPD resulted in apoptosis and mitochondrial injury, as corroborated by analyses using transmission electron microscopy and flow cytometry. Significantly, our study demonstrated the activation of the autophagy pathway in both the heart of zebrafish and cardiomyocytes, as substantiated by transmission electron microscopy and immunofluorescent techniques. Importantly, the increased the expression of P62 in the heart and cardiomyocytes suggested an inhibition of the autophagic process. The reduction in autophagy flux was also verified through in vivo experiments involving the infection of mCherry-GFP-LC3. We further identified that the fusion of autophagosomes and lysosomes was impaired in the 6PPD treatment group. In summary, our findings indicated that the impaired fusion of autophagosomes and lysosomes hampered the autophagic degradation process, leading to apoptosis and ultimately resulting in cardiac dysfunction and myocardial injury. This study discovered the crucial role of the autophagy pathway in regulating 6PPD-induced cardiotoxicity. SYNOPSIS: 6PPD exposure inhibited the autophagic degradation process and induced mitochondrial injury and apoptosis in the heart of adult zebrafish.
Collapse
Affiliation(s)
- Chanlin Fang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China
| | - Shanshan Di
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China
| | - Yundong Yu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China; State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China
| | - Peipei Qi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China
| | - Xinquan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, PR China.
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, China.
| |
Collapse
|
12
|
Li J, Xie Y, Zheng S, He H, Wang Z, Li X, Jiao S, Liu D, Yang F, Zhao H, Li P, Sun Y. Targeting autophagy in diabetic cardiomyopathy: From molecular mechanisms to pharmacotherapy. Biomed Pharmacother 2024; 175:116790. [PMID: 38776677 DOI: 10.1016/j.biopha.2024.116790] [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: 03/10/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 05/25/2024] Open
Abstract
Diabetic cardiomyopathy (DCM) is a cardiac microvascular complication caused by metabolic disorders. It is characterized by myocardial remodeling and dysfunction. The pathogenesis of DCM is associated with abnormal cellular metabolism and organelle accumulation. Autophagy is thought to play a key role in the diabetic heart, and a growing body of research suggests that modulating autophagy may be a potential therapeutic strategy for DCM. Here, we have summarized the major signaling pathways involved in the regulation of autophagy in DCM, including Adenosine 5'-monophosphate-activated protein kinase (AMPK), mechanistic target of rapamycin (mTOR), Forkhead box subfamily O proteins (FOXOs), Sirtuins (SIRTs), and PTEN-inducible kinase 1 (PINK1)/Parkin. Given the significant role of autophagy in DCM, we further identified natural products and chemical drugs as regulators of autophagy in the treatment of DCM. This review may help to better understand the autophagy mechanism of drugs for DCM and promote their clinical application.
Collapse
Affiliation(s)
- Jie Li
- China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Beijing, China
| | - Yingying Xie
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuwen Zheng
- Beijing University of Chinese Medicine School of Traditional Chinese Medicine, Beijing, China
| | - Haoming He
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhe Wang
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xuexi Li
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Siqi Jiao
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Dong Liu
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China
| | - Furong Yang
- Beijing University of Chinese Medicine School of Traditional Chinese Medicine, Beijing, China
| | - Hailing Zhao
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.
| | - Ping Li
- Beijing Key Laboratory for Immune-Mediated Inflammatory Diseases, Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.
| | - Yihong Sun
- Department of Cardiology, China-Japan Friendship Hospital (Institute of Clinical Medical Sciences), Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing, China.
| |
Collapse
|
13
|
Hu R, Huang Y, Jiang X, Xu Y, Zheng Z, Shi Y, Liu Y. Maternal dietary copper deficiency induces cardiomyopathy and liver injury in mice by activating autophagy. Nutr Res 2024; 126:1-10. [PMID: 38555686 DOI: 10.1016/j.nutres.2024.02.010] [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: 09/20/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 04/02/2024]
Abstract
Maternal nutritional deficiencies during pregnancy result in birth defects and elevate the risk of cardiovascular diseases and metabolic diseases. Accumulating evidence suggests that deficiency of copper, a fundamental trace element involved in several pivotal physiological processes, promotes the onset of multiple diseases, notably heart and liver diseases. Yet, exploration into the effects of maternal copper deficiency (CuD) on offspring is still limited. In this study, we hypothesized that maternal CuD induced cardiomyopathy and liver injury in offspring through the activation of autophagy. We established a maternal CuD mouse model by feeding pregnant C57BL/6 mice with a CuD diet until the end of the experiment. Echocardiography, histological analysis, western blotting, and quantitative polymerase chain reaction were performed on offspring at postnatal day 14. We found that maternal CuD caused growth retardation and early postnatal death in the offspring. Furthermore, our results revealed that CuD induced cardiac systolic dysfunction, cardiac hypertrophy, hepatic steatosis, and liver injury. Moreover, higher expression of LC3 and lower expression of p62 were observed in the heart tissues and liver tissues of CuD mice compared with the control group, indicating that CuD induced autophagy activation. In conclusion, maternal CuD caused severely deleterious effects on the heart and liver of the offspring via activating autophagy.
Collapse
Affiliation(s)
- Ruixiang Hu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Jinan University, Guangzhou 510630, PR China
| | - Yipu Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Xin Jiang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yi Xu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Zhirui Zheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yuansen Shi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yun Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
14
|
Li WW, Guo ZM, Wang BC, Liu QQ, Zhao WA, Wei XL. PCSK9 induces endothelial cell autophagy by regulating the PI3K/ATK pathway in atherosclerotic coronary heart disease. Clin Hemorheol Microcirc 2024:CH242172. [PMID: 38728182 DOI: 10.3233/ch-242172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
OBJECTIVE Atherosclerosis is a chronic inflammatory disease of the arteries, and its pathogenesis is related to endothelial dysfunction. It has been found that the protein convertase subtilin/kexin9 type (PCSK9) plays an important role in AS, but its specific mechanism is still unclear. METHODS In this study, we first cultured human umbilical vein endothelial cells (HUVECs) with 50 or 100μg/ml oxidized low-density lipoprotein (ox-LDL) for 24 hours to establish a coronary atherosclerosis cell model. RESULTS The results showed that ox-LDL induced HUVEC injury and autophagy and upregulated PCSK9 protein expression in HUVECs in a concentration-dependent manner. Silencing PCSK9 expression with siRNA inhibited ox-LDL-induced HUVEC endothelial dysfunction, inhibited the release of inflammatory factors, promoted HUVEC proliferation and inhibited apoptosis. In addition, ox-LDL increased the expression of LC3B-I and LC3B-II and decreased the expression of p62. However, these processes are reversed by sh-PCSK9. In addition, sh-PCSK9 can inhibit PI3K, AKT and mTOR phosphorylation and promote autophagy. CONCLUSION Taken together, our research shows that silencing PCSK9 inhibits the PI3K/ATK/mTOR pathway to activate ox-LDL-induced autophagy in vascular endothelial cells, alleviating endothelial cell injury and inflammation.
Collapse
Affiliation(s)
- Wei-Wei Li
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Ze-Ming Guo
- Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Bing-Cai Wang
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qing-Quan Liu
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wen-An Zhao
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiao-Lan Wei
- Department of Neurology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| |
Collapse
|
15
|
Cui X, Wang B, Han D, Cheng M, Yuan P, Du P, Hou Y, Su C, Tang J, Zhang J. Exacerbation of atherosclerosis by STX17 knockdown: Unravelling the role of autophagy and inflammation. J Cell Mol Med 2024; 28:e18402. [PMID: 39008328 PMCID: PMC11133389 DOI: 10.1111/jcmm.18402] [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: 11/24/2023] [Revised: 04/13/2024] [Accepted: 04/27/2024] [Indexed: 07/16/2024] Open
Abstract
Syntaxin 17 (STX17) has been identified as a crucial factor in mediating the fusion of autophagosomes and lysosomes. However, its specific involvement in the context of atherosclerosis (AS) remains unclear. This study sought to elucidate the role and mechanistic contributions of STX17 in the initiation and progression of AS. Utilizing both in vivo and in vitro AS model systems, we employed ApoE knockout (KO) mice subjected to a high-fat diet and human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL) to assess STX17 expression. To investigate underlying mechanisms, we employed shRNA-STX17 lentivirus to knock down STX17 expression, followed by evaluating autophagy and inflammation in HUVECs. In both in vivo and in vitro AS models, STX17 expression was significantly upregulated. Knockdown of STX17 exacerbated HUVEC damage, both with and without ox-LDL treatment. Additionally, we observed that STX17 knockdown impaired autophagosome degradation, impeded autophagy flux and also resulted in the accumulation of dysfunctional lysosomes in HUVECs. Moreover, STX17 knockdown intensified the inflammatory response following ox-LDL treatment in HUVECs. Further mechanistic exploration revealed an association between STX17 and STING; reducing STX17 expression increased STING levels. Further knockdown of STING enhanced autophagy flux. In summary, our findings suggest that STX17 knockdown worsens AS by impeding autophagy flux and amplifying the inflammatory response. Additionally, the interaction between STX17 and STING may play a crucial role in STX17-mediated autophagy.
Collapse
Affiliation(s)
- Xinyue Cui
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Bo Wang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Dongjian Han
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Mengdie Cheng
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Peiyu Yuan
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Pengchong Du
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Yachen Hou
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Chang Su
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Junnan Tang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| | - Jinying Zhang
- Department of CardiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardiac Injury and Repair of Henan ProvinceZhengzhouHenanChina
| |
Collapse
|
16
|
Sniezek Carney O, Harris KW, Wohlfarter Y, Lee K, Butschek G, Anzmann A, Claypool SM, Hamacher-Brady A, Keller M, Vernon HJ. Stem cell models of TAFAZZIN deficiency reveal novel tissue-specific pathologies in Barth Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.28.591534. [PMID: 38746168 PMCID: PMC11092433 DOI: 10.1101/2024.04.28.591534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Barth syndrome (BTHS) is a rare mitochondrial disease caused by pathogenic variants in the gene TAFAZZIN, which leads to abnormal cardiolipin (CL) metabolism on the inner mitochondrial membrane. Although TAFAZZIN is ubiquitously expressed, BTHS involves a complex combination of tissue specific phenotypes including cardiomyopathy, neutropenia, skeletal myopathy, and growth delays, with a relatively minimal neurological burden. To understand both the developmental and functional effects of TAZ-deficiency in different tissues, we generated isogenic TAZ knockout (TAZ- KO) and WT cardiomyocytes (CMs) and neural progenitor cells (NPCs) from CRISPR-edited induced pluripotent stem cells (iPSCs). In TAZ-KO CMs we discovered evidence of dysregulated mitophagy including dysmorphic mitochondria and mitochondrial cristae, differential expression of key autophagy-associated genes, and an inability of TAZ-deficient CMs to properly initiate stress-induced mitophagy. In TAZ-deficient NPCs we identified novel phenotypes including a reduction in CIV abundance and CIV activity in the CIII2&CIV2 intermediate complex. Interestingly, while CL acyl chain manipulation was unable to alter mitophagy defects in TAZ-KO CMs, we found that linoleic acid or oleic acid supplementation was able to partially restore CIV abundance in TAZ-deficient NPCs. Taken together, our results have implications for understanding the tissue-specific pathology of BTHS and potential for tissue-specific therapeutic targeting. Moreover, our results highlight an emerging role for mitophagy in the cardiac pathophysiology of BTHS and reveal a potential neuron-specific bioenergetic phenotype.
Collapse
|
17
|
Li Y, Liu Y, Yao X, Zhu L, Yang L, Zhan Q. MLKL Protects Pulmonary Endothelial Cells in Acute Lung Injury. Am J Respir Cell Mol Biol 2024; 70:295-307. [PMID: 38207123 DOI: 10.1165/rcmb.2023-0207oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 01/10/2024] [Indexed: 01/13/2024] Open
Abstract
The role of autophagy in pulmonary microvascular endothelial cells (PMVECs) is controversial in LPS-induced acute lung injury (ALI). Mixed lineage kinase domain-like pseudokinase (MLKL) has recently been reported to maintain cell survival by facilitating autophagic flux in response to starvation rather than its well-recognized role in necroptosis. Using a mouse PMVEC and LPS-induced ALI model, we showed that in PMVECs, MLKL was phosphorylated (p-MLKL) and autophagic flux was accelerated at the early stage of LPS stimulation (1-3 h), manifested by increases in concentrations of lipidated MAP1LC3B/LC3B (microtubule-associated protein 1 light chain 3 β; LC3-II), decreases in concentrations of SQSTM1/p62 (sequestosome 1), and fusion of the autophagosome and lysosome by pHluorin-mKate2-human LC3 assay, which were all reversed by either MLKL inhibitor or siRNA MLKL. In mice, the inhibition of MLKL increased vascular permeability and aggravated mouse ALI upon 3-hour LPS stimulation. The p-MLKL induced by short-term LPS formed multimers to facilitate the closure of the phagophore by HaloTag-LC3 autophagosome completion assay. The charged multivesicular body protein 2A (CHMP2A) is essential in the process of phagophore closure into the nascent autophagosome. In agreement with the p-MLKL change, CHMP2A concentrations markedly increased during 1-3-hour LPS stimulation. CHMP2A knockdown blocked autophagic flux upon LPS stimulation, whereas CHMP2A overexpression boosted autophagic flux and attenuated mouse ALI even in the presence of MLKL inhibitor. We propose that the activated MLKL induced by short-term LPS facilitates autophagic flux by accelerating the closure of the phagophore via CHMP2A, thus protecting PMVECs and alleviating LPS-induced ALI.
Collapse
Affiliation(s)
- Ying Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; and
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Yingxiang Liu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; and
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Xueya Yao
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; and
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Ling Zhu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; and
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Liqun Yang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; and
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| | - Qionghui Zhan
- Department of Anesthesiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine; and
- Key Laboratory of Anesthesiology, Shanghai Jiao Tong University, Ministry of Education, Shanghai, China
| |
Collapse
|
18
|
Ma Y, Zhao HP, Yang LG, Li L, Wang AL, Zhang XJ, Wang K, Yang B, Zhu ZF, Zhang PJ, Wang JP, Chi RF, Li B, Qin FZ, Wang ZP. NADPH oxidase 2 mediates cardiac sympathetic denervation and myocyte autophagy, resulting in cardiac atrophy and dysfunction in doxorubicin-induced cardiomyopathy. Sci Rep 2024; 14:6971. [PMID: 38521855 PMCID: PMC10960835 DOI: 10.1038/s41598-024-57090-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 03/14/2024] [Indexed: 03/25/2024] Open
Abstract
Doxorubicin has been used extensively as a potent anticancer agent, but its clinical use is limited by its cardiotoxicity. However, the underlying mechanisms remain to be fully elucidated. In this study, we tested whether NADPH oxidase 2 (Nox2) mediates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy, resulting in cardiac atrophy and dysfunction in doxorubicin-induced heart failure. Nox2 knockout (KO) and wild-type (WT) mice were randomly assigned to receive a single injection of doxorubicin (15 mg/kg, i.p.) or saline. WT doxorubicin mice exhibited the decreases in survival rate, left ventricular (LV) wall thickness and LV fractional shortening and the increase in the lung wet-to-dry weight ratio 1 week after the injections. These alterations were attenuated in Nox2 KO doxorubicin mice. In WT doxorubicin mice, myocardial oxidative stress was increased, myocardial noradrenergic nerve fibers were reduced, myocardial expression of PGP9.5, GAP43, tyrosine hydroxylase and norepinephrine transporter was decreased, and these changes were prevented in Nox2 KO doxorubicin mice. Myocyte autophagy was increased and myocyte size was decreased in WT doxorubicin mice, but not in Nox2 KO doxorubicin mice. Nox2 mediates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy-both of which contribute to cardiac atrophy and failure after doxorubicin treatment.
Collapse
Affiliation(s)
- Yuan Ma
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Hui-Ping Zhao
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Li-Guo Yang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Lu Li
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Ai-Lin Wang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Xiao-Juan Zhang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Ke Wang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Bin Yang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Zong-Feng Zhu
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Pei-Jun Zhang
- Shanxi Datong University School of Medicine, Datong, 037009, Shanxi, People's Republic of China
| | - Jia-Pu Wang
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Rui-Fang Chi
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Bao Li
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China
| | - Fu-Zhong Qin
- The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, People's Republic of China.
- Shanxi Medical University, Taiyuan, 030001, Shanxi, People's Republic of China.
| | - Zhi-Peng Wang
- Institute for Radiation Protection, Taiyuan, 030006, Shanxi, People's Republic of China
| |
Collapse
|
19
|
Zhao HP, Ma Y, Zhang XJ, Guo HX, Yang B, Chi RF, Zhang NP, Wang JP, Li B, Qin FZ, Yang LG. NADPH oxidase 2 inhibitor GSK2795039 prevents doxorubicin-induced cardiac atrophy by attenuating cardiac sympathetic nerve terminal abnormalities and myocyte autophagy. Eur J Pharmacol 2024; 967:176351. [PMID: 38290568 DOI: 10.1016/j.ejphar.2024.176351] [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: 09/08/2023] [Revised: 12/19/2023] [Accepted: 01/22/2024] [Indexed: 02/01/2024]
Abstract
Doxorubicin is widely used for the treatment of human cancer, but its clinical use is limited by a cumulative dose-dependent cardiotoxicity. However, the mechanism of doxorubicin-induced cardiac atrophy and failure remains to be fully understood. In this study, we tested whether the specific NADPH oxidase 2 (Nox2) inhibitor GSK2795039 attenuates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy, leading to the amelioration of cardiac atrophy and dysfunction in chronic doxorubicin-induced cardiomyopathy. Mice were randomized to receive saline, doxorubicin (2.5 mg/kg, every other day, 6 times) or doxorubicin plus GSK2795039 (2.5 mg/kg, twice a day, 9 weeks). Left ventricular (LV) total wall thickness and LV ejection fraction were decreased in doxorubicin-treated mice compared with saline-treated mice and the decreases were prevented by the treatment of the specific Nox2 inhibitor GSK2795039. The ratio of total heart weight to tibia length and myocyte cross-sectional area were decreased in doxorubicin-treated mice, and the decreases were attenuated by the GSK2795039 treatment. In doxorubicin-treated mice, myocardial Nox2 and 4-hydroxynonenal levels were increased, myocardial expression of GAP43, tyrosine hydroxylase and norepinephrine transporter, markers of sympathetic nerve terminals, was decreased, and these changes were prevented by the GSK2795039 treatment. The ratio of LC3 II/I, a marker of autophagy, and Atg5, Atg12 and Atg12-Atg5 conjugate proteins were increased in doxorubicin-treated mice, and the increases were attenuated by the GSK2795039 treatment. These findings suggest that inhibition of Nox2 by GSK2795039 attenuates cardiac sympathetic nerve terminal abnormalities and myocyte autophagy, thereby ameliorating cardiac atrophy and dysfunction after chronic doxorubicin treatment.
Collapse
Affiliation(s)
- Hui-Ping Zhao
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Yuan Ma
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Xiao-Juan Zhang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Hong-Xia Guo
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Bin Yang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Rui-Fang Chi
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Nian-Ping Zhang
- Shanxi Datong University School of Medicine, Datong, 037009, Shanxi, PR China
| | - Jia-Pu Wang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Bao Li
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China
| | - Fu-Zhong Qin
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China.
| | - Li-Guo Yang
- The Second Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Medical University, Taiyuan, 030001, Shanxi, PR China; Shanxi Provincial People's Hospital, Taiyuan, 030001, Shanxi, PR China
| |
Collapse
|
20
|
Grazide MH, Ruidavets JB, Martinet W, Elbaz M, Vindis C. Association of Circulating Autophagy Proteins ATG5 and Beclin 1 with Acute Myocardial Infarction in a Case-Control Study. Cardiology 2024; 149:217-224. [PMID: 38432214 PMCID: PMC11152019 DOI: 10.1159/000537816] [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: 11/27/2023] [Accepted: 02/09/2024] [Indexed: 03/05/2024]
Abstract
INTRODUCTION Acute myocardial infarction (AMI) is a main contributor of sudden cardiac death worldwide. The discovery of new biomarkers that can improve AMI risk prediction meets a major clinical need for the identification of high-risk patients and the tailoring of medical treatment. Previously, we reported that autophagy a highly conserved catabolic mechanism for intracellular degradation of cellular components is involved in atherosclerotic plaque phenotype and cardiac pathological remodeling. The crucial role of autophagy in the normal and diseased heart has been well described, and its activation functions as a pro-survival process in response to myocardial ischemia. However, autophagy is dysregulated in ischemia/reperfusion injury, thus promoting necrotic or apoptotic cardiac cell death. Very few studies have focused on the plasma levels of autophagy markers in cardiovascular disease patients, even though they could be companion biomarkers of AMI injury. The aims of the present study were to evaluate (1) whether variations in plasma levels of two key autophagy regulators autophagy-related gene 5 (ATG5) and Beclin 1 (the mammalian yeast ortholog Atg6/Vps30) are associated with AMI and (2) their potential for predicting AMI risk. METHODS The case-control study population included AMI patients (n = 100) and control subjects (n = 99) at high cardiovascular risk but without known coronary disease. Plasma levels of ATG5 and Beclin 1 were measured in the whole population study by enzyme-linked immunosorbent assay. RESULTS Multivariate analyses adjusted on common cardiovascular factors and medical treatments, and receiver operating characteristic curves demonstrated that ATG5 and Beclin 1 levels were inversely associated with AMI and provided original biomarkers for AMI risk prediction. CONCLUSION Plasma levels of autophagy regulators ATG5 and Beclin 1 represent relevant candidate biomarkers associated with AMI.
Collapse
Affiliation(s)
- Marie-Hélène Grazide
- Center for Clinical Investigation (CIC1436)/CARDIOMET, Rangueil University Hospital, Toulouse, France
- University of Toulouse III, Toulouse, France
| | | | - Wim Martinet
- Laboratory of Physiopharmacology, University of Antwerp, Antwerp, Belgium
| | - Meyer Elbaz
- Center for Clinical Investigation (CIC1436)/CARDIOMET, Rangueil University Hospital, Toulouse, France
- University of Toulouse III, Toulouse, France
- Department of Cardiology, Rangueil University Hospital, Toulouse, France
| | - Cécile Vindis
- Center for Clinical Investigation (CIC1436)/CARDIOMET, Rangueil University Hospital, Toulouse, France
- University of Toulouse III, Toulouse, France
| |
Collapse
|
21
|
Wang D, Wang D, Jin Q, Wang X. Suxiao Jiuxin Pill alleviates myocardial ischemia/reperfusion-induced autophagy via miR-193a-3p/ALKBH5 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 125:155359. [PMID: 38301300 DOI: 10.1016/j.phymed.2024.155359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Myocardial ischemia/reperfusion injury (MIRI) poses a formidable challenge to cardiac reperfusion therapy due to the absence of effective clinical interventions. Methylation of N6-methyladenosine (m6A), which is the most common post-transcriptional modifications occurring within mammalian mRNA, is believed to be involved in MIRI by modulating autophagy. MicroRNAs (miRNAs) play a crucial role in regulating gene expression at the post-transcriptional level and have been implicated in the regulation of m6A methylation. Suxiao Jiuxin Pill (SJP) is extensively used in China for the clinical treatment of angina pectoris and confers benefits to patients with acute coronary syndrome who have received percutaneous coronary intervention. However, the precise mechanisms underlying SJP intervention in MIRI remain unclear. PURPOSE This study aimed to demonstrate, both in vivo and in vitro, that SJP could alleviate autophagy in MIRI by regulating miR-193a-3p to target and upregulate the demethylase ALKBH5. METHODS An in vitro hypoxia/reoxygenation model was established using H9c2 cells, while an in vivo MIRI model was established using Wistar rats. A lentivirus harboring the precursor sequence of miR-193a-3p was employed for its overexpression. Adeno-associated viruses were used to silence both miR-193a-3p and ALKBH5 expressions. Cardiac function, infarct size, and tissue structure in rats were assessed using echocardiography, triphenyl tetrazolium chloride (TTC) staining, and HE staining, respectively. Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) was employed to detect the levels of apoptosis in rat cardiac tissue. m6A methylation levels were assessed using colorimetry. GFP-RFP-LC3B was used to monitor autophagic flux and transmission electron microscopy was used to evaluate the development of autophagosomes. Western Blot and qRT-PCR were respectively employed to assess the levels of autophagy-related proteins and miR-193a-3p. RESULTS SJP alleviated autophagy, preserved cardiac function, and minimized myocardial damage in the hearts of MIRI rats. SJP attenuated autophagy in H/R H9C2 cells. Elevated levels of miR-193a-3p were observed in the cardiac tissues of MIRI rats and H/R H9C2 cells, whereas SJP downregulated miR-193a-3p levels in these models. ALKBH5, a target gene of miR-193, is negatively regulated by miR-193a-3p. Upon overexpression of miR-193a-3p or silencing of ALKBH5, m6A methylation decreased, and the autophagy-attenuating effects of SJP and its components, senkyunolide A and l-borneol, were lost in H/R H9C2 cells, whereas in MIRI rats, these effects were not abolished but merely weakened. Further investigation indicated that the METTL3 inhibitor STM2475, combined with the silencing of miR-193a-3p, similarly attenuated autophagy in the hearts of MIRI rats. This suggests that a reduction in m6A methylation is involved in autophagy alleviation. CONCLUSION We demonstrated that SJP mitigates autophagy in MIRI by downregulating miR-193a-3p, enhancing ALKBH5 expression, and reducing m6A methylation, a mechanism potentially attributed to its constituents, senkyunolide A and l-borneol.
Collapse
Affiliation(s)
- Dongyuan Wang
- Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Wang
- Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qipeng Jin
- Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaolong Wang
- Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China; Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
22
|
Xu J, Chen F, Zhu W, Zhang W. NPC1 promotes autophagy with tumor promotion and acts as a prognostic model for hepatocellular carcinoma. Gene 2024; 897:148050. [PMID: 38042211 DOI: 10.1016/j.gene.2023.148050] [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: 08/16/2023] [Revised: 11/02/2023] [Accepted: 11/29/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND more and more studies have indicated that autophagy plays a crucial role in hepatocellular carcinoma (HCC) in recent years. Hence, our study aimed to establish a prognostic signature for HCC based on autophagy-related genes (ARGs) in order to predict the prognosis of HCC. METHODS All original gene-expression data and clinical information were downloaded from The Cancer Genome Atlas (TCGA), International Cancer Genome Consortium (ICGC) and Gene Expression Omnibus (GEO). ARGs were obtained from the Human Autophagy Database (HADb). Univariate Cox regression analysis, Least absolute shrinkage and selection operator (LASSO) and Principal Component Analysis (PCA) analysis were performed to identify and validate the validity and reliability of our eight-gene signature, Gene Set Enrichment Analysis (GSEA) was used to perform enrichment analysis by comparing high-risk and low-risk groups in KEGG (Kyoto Encyclopedia of Genes and Genomes) and GO (Gene Ontology) gene sets. Finally, we verified the gene (NPC1) by functional experiments in vitro and in vivo. RESULTS 8 ARGs were identified for establishing an eight-gene signature. Then, we validated our eight-gene signature in training, internal, external, and entire testing cohorts. Besides, we also explored the relationships between the eight-gene signature and immune infiltration or immune checkpoints. We also identified NPC1 was closely related to Activated CD4 T cell and Type I IFN Response, and higher expressed level of HCC patients was more sensitive to CTLA4 and TNFRSF9 immune checkpoint inhibitors. NPC1 is highly expressed in HCC cells and tumor tissues, which promotes the proliferation, migration, and invasion of tumor cells by activating autophagy.. CONCLUSION 8 ARGs were used to establish a gene signature to predict the prognosis of HCC. we inferred that NPC1 can promote late autophagy, it could be a future novel therapeutic target of HCC.
Collapse
Affiliation(s)
- Jian Xu
- Department of Hepatobiliary Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Fei Chen
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Wenjie Zhu
- Department of Gastrointestinal Surgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Wei Zhang
- Department of Nuclear Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
23
|
Xu Z, Pan Z, Jin Y, Gao Z, Jiang F, Fu H, Chen X, Zhang X, Yan H, Yang X, Yang B, He Q, Luo P. Inhibition of PRKAA/AMPK (Ser485/491) phosphorylation by crizotinib induces cardiotoxicity via perturbing autophagosome-lysosome fusion. Autophagy 2024; 20:416-436. [PMID: 37733896 PMCID: PMC10813574 DOI: 10.1080/15548627.2023.2259216] [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: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/23/2023] Open
Abstract
Crizotinib, a small-molecule tyrosine kinase inhibitor targeting ALK, MET and ROS1, is the first-line drug for ALK-positive metastatic non-small cell lung cancer and is associated with severe, sometimes fatal, cases of cardiac failure, which increases the risk of mortality. However, the underlying mechanism remains unclear, which causes the lack of therapeutic strategy. We established in vitro and in vivo models for crizotinib-induced cardiotoxicity and found that crizotinib caused left ventricular dysfunction, myocardial injury and pathological remodeling in mice and induced cardiomyocyte apoptosis and mitochondrial injury. In addition, we found that crizotinib prevented the degradation of MET protein by interrupting autophagosome-lysosome fusion and silence of MET or re-activating macroautophagy/autophagy flux rescued the cardiomyocytes death and mitochondrial injury caused by crizotinib, suggesting that impaired autophagy activity is the key reason for crizotinib-induced cardiotoxicity. We further confirmed that recovering the phosphorylation of PRKAA/AMPK (Ser485/491) by metformin re-activated autophagy flux in cardiomyocytes and metformin rescued crizotinib-induced cardiomyocyte injury and cardiac complications. In summary, we revealed a novel mechanism for crizotinib-induced cardiotoxicity, wherein the crizotinib-impaired autophagy process causes cardiomyocyte death and cardiac injury by inhibiting the degradation of MET protein, demonstrated a new function of impeded autophagosome-lysosome fusion in drugs-induced cardiotoxicity, pointed out the essential role of the phosphorylation of PRKAA (Ser485/491) in autophagosome-lysosome fusion and confirmed metformin as a potential therapeutic strategy for crizotinib-induced cardiotoxicity.Abbreviations and Acronyms: AAV: adeno-associated virus; ACAC/ACC: acetyl-Co A carboxylase; AMP: adenosine monophosphate; AMPK: AMP-activated protein kinase; ATG5: autophagy related 5; ATG7: autophagy related 7; CHX: cycloheximide; CKMB: creatine kinase myocardial band; CQ: chloroquine; c-PARP: cleaved poly (ADP-ribose) polymerase; DAPI: 4'6-diamidino-2-phenylindole; EF: ejection fraction; FOXO: forkhead box O; FS: fractional shortening; GSEA: gene set enrichment analysis; H&E: hematoxylin and eosin; HF: heart failure; HW: TL: ratio of heart weight to tibia length; IR: ischemia-reperfusion; KEGG: Kyoto encyclopedia of genes and genomes; LAMP2: lysosomal-associated membrane protein 2; LDH: lactate dehydrogenase; MCMs: mouse cardiomyocytes; MMP: mitochondrial membrane potential; mtDNA: mitochondrial DNA; MYH6: myosin, heavy peptide 6, cardiac muscle, alpha; MYH7: myosin, heavy peptide 7, cardiac muscle, beta; NPPA: natriuretic peptide type A; NPPB: natriuretic peptide type B; PI: propidium iodide; PI3K: phosphoinositide 3-kinase; PRKAA/AMPKα: protein kinase AMP-activated catalytic subunit alpha; qPCR: quantitative real-time PCR; SD: standard deviation; SRB: sulforhodamine B; TKI: tyrosine kinase inhibitor; WGA: wheat germ agglutinin.
Collapse
Affiliation(s)
- Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Zezheng Pan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Ying Jin
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Zizheng Gao
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Feng Jiang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Huangxi Fu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Xueqin Chen
- Department of Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R.China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Xiaochen Zhang
- Department of Medical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R.China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R.China
- Deparment of Pharmaceutical and Translational Toxicology, Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, P.R.China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
- Department of Cardiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, P.R.China
- Department of Pharmacology and Toxicology, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, P.R.China
| |
Collapse
|
24
|
Zhang Q, Li Z, Li Q, Trammell SA, Schmidt MS, Pires KM, Cai J, Zhang Y, Kenny H, Boudina S, Brenner C, Abel ED. Control of NAD + homeostasis by autophagic flux modulates mitochondrial and cardiac function. EMBO J 2024; 43:362-390. [PMID: 38212381 PMCID: PMC10897141 DOI: 10.1038/s44318-023-00009-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 10/31/2023] [Accepted: 11/08/2023] [Indexed: 01/13/2024] Open
Abstract
Impaired autophagy is known to cause mitochondrial dysfunction and heart failure, in part due to altered mitophagy and protein quality control. However, whether additional mechanisms are involved in the development of mitochondrial dysfunction and heart failure in the setting of deficient autophagic flux remains poorly explored. Here, we show that impaired autophagic flux reduces nicotinamide adenine dinucleotide (NAD+) availability in cardiomyocytes. NAD+ deficiency upon autophagic impairment is attributable to the induction of nicotinamide N-methyltransferase (NNMT), which methylates the NAD+ precursor nicotinamide (NAM) to generate N-methyl-nicotinamide (MeNAM). The administration of nicotinamide mononucleotide (NMN) or inhibition of NNMT activity in autophagy-deficient hearts and cardiomyocytes restores NAD+ levels and ameliorates cardiac and mitochondrial dysfunction. Mechanistically, autophagic inhibition causes the accumulation of SQSTM1, which activates NF-κB signaling and promotes NNMT transcription. In summary, we describe a novel mechanism illustrating how autophagic flux maintains mitochondrial and cardiac function by mediating SQSTM1-NF-κB-NNMT signaling and controlling the cellular levels of NAD+.
Collapse
Affiliation(s)
- Quanjiang Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Zhonggang Li
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Department of Human Genetics, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Qiuxia Li
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Samuel Aj Trammell
- Department of Biomedical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Mark S Schmidt
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Karla Maria Pires
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Jinjin Cai
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
| | - Yuan Zhang
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Helena Kenny
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Sihem Boudina
- Division of Endocrinology, Metabolism and Diabetes, and Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, UT, 84112, USA
- Department of Nutrition and Integrative Physiology, College of Health, University of Utah, Salt Lake City, UT, 84112, USA
| | - Charles Brenner
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Department of Diabetes & Cancer Metabolism, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - E Dale Abel
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, David Geffen School of Medicine and UCLA Health, University of California-Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA.
| |
Collapse
|
25
|
Peng Y, Liao B, Zhou Y, Zeng W. Ginsenoside Rb2 improves heart failure by down-regulating miR-216a-5p to promote autophagy and inhibit apoptosis and oxidative stress. J Appl Biomed 2023; 21:180-192. [PMID: 38112457 DOI: 10.32725/jab.2023.024] [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: 03/02/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Ginsenoside Rb2 is beneficial in cardiovascular disease treatment, yet its role in heart failure (HF) is obscure. This study aimed to investigate the effect and mechanism of ginsenoside Rb2 on HF. METHODS The left anterior descending branch-ligated HF rat model and oxygen-glucose deprivation/reoxygenation (OGD/R) H9c2 cell model were constructed. Ginsenoside Rb2 were applied for intervention. Heart function indexes, miR-216a-5p expression, autophagy, oxidative stress, apoptosis, cell morphology, and proliferation were detected to explore the effect of ginsenoside Rb2 on HF. Overexpression of miR-216a-5p was employed to explore the specific mechanism of ginsenoside Rb2 on HF. RESULTS Ginsenoside Rb2 improved the heart function of HF rats, including the reduction of heart rate, LVEDP, and heart weight/body weight ratio, and the increase of LVSP, +dP/dtmax, -dP/dtmax, LVEF, and LVFS. It also down-regulated miR-216a-5p expression and enhanced OGD/R-induced cardiomyocyte viability. Ginsenoside Rb2 up-regulated Bcl2, LC3B II/I, and Beclin1, and down-regulated Bax, Caspase-3, and p62 in the myocardium of HF rats and OGD/R-induced H9c2 cells. Moreover, ginsenoside Rb2 increased the levels of SOD and CAT, but decreased the levels of MDA and ROS in the myocardium of HF rats and OGD/R-induced H9c2 cells. However, overexpression of miR-216a-5p promoted the apoptosis and oxidative stress of cardiomyocytes and inhibited autophagy, thus reversing the therapeutic effect of ginsenoside Rb2 on HF in vivo and in vitro. CONCLUSION Ginsenoside Rb2 demonstrated potential as a therapeutic intervention for HF by enhancing autophagy and reducing apoptosis and oxidative stress through miR-216a-5p downregulation. Further research could explore its application in clinical trials and investigate the complex mechanism networks underlying its effects.
Collapse
Affiliation(s)
- You Peng
- The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Center of Geriatric, Changsha, Hunan, China
- Hunan Research Institute of Geriatrics, Changsha, Hunan, China
- Major Chronic Disease Research Center of Hunan Provincial Geriatric Institute, Changsha, Hunan, China
| | - Bin Liao
- The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Center of Geriatric, Changsha, Hunan, China
| | - Yan Zhou
- The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Center of Geriatric, Changsha, Hunan, China
| | - Wei Zeng
- The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Center of Geriatric, Changsha, Hunan, China
| |
Collapse
|
26
|
Xuan C, Hu R. Chemical Biology Perspectives on STING Agonists as Tumor Immunotherapy. ChemMedChem 2023; 18:e202300405. [PMID: 37794702 DOI: 10.1002/cmdc.202300405] [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: 08/01/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/06/2023]
Abstract
Stimulator of interferon genes (STING) is a crucial adaptor protein in the innate immune response. STING activation triggers cytokine secretion, including type I interferon and initiates T cell-mediated adaptive immunity. The activated immune system converts "cold tumors" into "hot tumors" that are highly responsive to T cells by recruiting them to the tumor microenvironment, ultimately leading to potent and long-lasting antitumor effects. Unlike most immune checkpoint inhibitors, STING agonists represent a groundbreaking class of innate immune agonists that hold great potential for effectively targeting various cancer populations and are poised to become a blockbuster in tumor immunotherapy. This review will focus on the correlation between the STING signaling pathway and tumor immunity, as well as explore the impact of STING activation on other biological processes. Ultimately, we will summarize the development and optimization of STING agonists from a medicinal chemistry perspective, evaluate their potential in cancer therapy, and identify possible challenges for future advancement.
Collapse
Affiliation(s)
- Chenyuan Xuan
- Department of Pharmacology, China Pharmaceutical University, No 24, TongJiaXiang, Gulou District, Nanjing, 210009, P. R. China
| | - Rong Hu
- Department of Pharmacology, China Pharmaceutical University, No 24, TongJiaXiang, Gulou District, Nanjing, 210009, P. R. China
| |
Collapse
|
27
|
Deng F, Xu P, Miao J, Jin C, Tu H, Zhang J. Pulmonary tuberculosis biomarker miR-215-5p inhibits autophagosome-lysosome fusion in macrophages. Tuberculosis (Edinb) 2023; 143:102422. [PMID: 37925930 DOI: 10.1016/j.tube.2023.102422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/17/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023]
Abstract
The normal autophagy flux is beneficial for the rapid elimination of phagocytic pathogens by macrophages. However, Mycobacterium tuberculosis interferes with the autophagy flux of macrophages to weaken their immune function and evade immune surveillance. In this study, we found that miRNA-215-5p was downregulated in tuberculosis patients. A potential diagnostic model for tuberculosis was established by combining miRNA-215-5p with three others differentially expressed microRNAs (miRNA-145-5p, miRNA-486-5p and miRNA-628-3p). Furthermore, we discovered that the up-regulated miRNA-215-5p could inhibit the maturation of autophagy by preventing the fusion of autophagosomes with lysosomes in macrophages. The role of TB-specific miRNA-215-5p in inhibiting auto-lysosome formation provides evidence of its potential role in Host-directed therapy for tuberculosis.
Collapse
Affiliation(s)
- Feng Deng
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Peng Xu
- Department of Clinical Laboratory, Affiliated Hospital of Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Jiahong Miao
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Cheng Jin
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China
| | - Huihui Tu
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China.
| | - Jianhua Zhang
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China.
| |
Collapse
|
28
|
Sklifasovskaya AP, Blagonravov M, Ryabinina A, Goryachev V, Syatkin S, Chibisov S, Akhmetova K, Prokofiev D, Agostinelli E. The role of heat shock proteins in the pathogenesis of heart failure (Review). Int J Mol Med 2023; 52:106. [PMID: 37772383 PMCID: PMC10558216 DOI: 10.3892/ijmm.2023.5309] [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: 07/11/2023] [Accepted: 09/11/2023] [Indexed: 09/30/2023] Open
Abstract
The influence of heat shock proteins (HSPs) on protein quality control systems in cardiomyocytes is currently under investigation. The effect of HSPs on the regulated cell death of cardiomyocytes (CMCs) is of great importance, since they play a major role in the implementation of compensatory and adaptive mechanisms in the event of cardiac damage. HSPs mediate a number of mechanisms that activate the apoptotic cascade, playing both pro‑ and anti‑apoptotic roles depending on their location in the cell. Another type of cell death, autophagy, can in some cases lead to cell death, while in other situations it acts as a cell survival mechanism. The present review considered the characteristics of the expression of HSPs of different molecular weights in CMCs in myocardial damage caused by heart failure, as well as their role in the realization of certain types of regulated cell death.
Collapse
Affiliation(s)
| | | | - Anna Ryabinina
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | | | - Sergey Syatkin
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Sergey Chibisov
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Karina Akhmetova
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Daniil Prokofiev
- Institute of Medicine, RUDN University, 117198 Moscow, Russia, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Faculty of Medicine and Dentistry, Sapienza University of Rome, University Hospital Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation, ETS-ONLUS, I-00159 Rome, Italy
| |
Collapse
|
29
|
Ye Q, Chen W, Fu H, Ding Y, Jing Y, Shen J, Yuan Z, Zha K. Targeting Autophagy in Atrial Fibrillation. Rev Cardiovasc Med 2023; 24:288. [PMID: 39077569 PMCID: PMC11273128 DOI: 10.31083/j.rcm2410288] [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: 11/17/2022] [Revised: 05/04/2023] [Accepted: 05/19/2023] [Indexed: 07/31/2024] Open
Abstract
Atrial fibrillation (AF) is the most common type of arrhythmia in clinical practice, and its incidence is positively correlated with risk factors that include advanced age, hypertension, diabetes, and heart failure. Although our understanding of the mechanisms that govern the occurrence and persistence of AF has been increasing rapidly, the exact mechanism of AF is still not fully understood. Autophagy is an evolutionarily highly conserved and specific physiological process in cells that has been suggested as a potential therapeutic target for several cardiovascular diseases including the pathophysiology of AF. The present article provides an updated review of the fast-progressing field of research surrounding autophagy in AF, and how regulating autophagy might be a therapeutic target to reduce the incidence of AF.
Collapse
Affiliation(s)
- Qiang Ye
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Wen Chen
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Hengsong Fu
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yanling Ding
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yuling Jing
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Jingsong Shen
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Ziyang Yuan
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Kelan Zha
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
30
|
Wei H, Yin Y, Yang W, Zhu J, Chen L, Guo R, Yang Z, Li S. Nuciferine induces autophagy to relieve vascular cell adhesion molecule 1 activation via repressing the Akt/mTOR/AP1 signal pathway in the vascular endothelium. Front Pharmacol 2023; 14:1264324. [PMID: 37841916 PMCID: PMC10569124 DOI: 10.3389/fphar.2023.1264324] [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: 07/20/2023] [Accepted: 09/14/2023] [Indexed: 10/17/2023] Open
Abstract
Pro-inflammatory factor-associated vascular cell adhesion molecule 1 (VCAM1) activation initiates cardiovascular events. This study aimed to explore the protective role of nuciferine on TNFα-induced VCAM1 activation. Nuciferine was administrated to both high-fat diet (HFD)-fed mice and the TNFα-exposed human vascular endothelial cell line. VCAM1 expression and further potential mechanism(s) were explored. Our data revealed that nuciferine intervention alleviated VCAM1 activation in response to both high-fat diet and TNFα exposure, and this protective effect was closely associated with autophagy activation since inhibiting autophagy by either genetic or pharmaceutical approaches blocked the beneficial role of nuciferine. Mechanistical studies revealed that Akt/mTOR inhibition, rather than AMPK, SIRT1, and p38 signal pathways, contributed to nuciferine-activated autophagy, which further ameliorated TNFα-induced VCAM1 via repressing AP1 activation, independent of transcriptional regulation by IRF1, p65, SP1, and GATA6. Collectively, our data uncovered a novel biological function for nuciferine in protecting VCAM1 activation, implying its potential application in improving cardiovascular events.
Collapse
Affiliation(s)
- Haibin Wei
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Biobank, Zhejiang Cancer Hospital, Hangzhou, Zhejiang, China
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yujie Yin
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Wenwen Yang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jinyan Zhu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lin Chen
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rui Guo
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhen Yang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Clinical Nutrition, Affiliated Zhejiang Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
31
|
Kolahdouzmohammadi M, Pahlavan S, Sotoodehnejadnematalahi F, Tahamtani Y, Totonchi M. Activation of AMPK promotes cardiac differentiation by stimulating the autophagy pathway. J Cell Commun Signal 2023; 17:939-955. [PMID: 37040028 PMCID: PMC10409960 DOI: 10.1007/s12079-023-00744-z] [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: 09/15/2022] [Accepted: 03/22/2023] [Indexed: 04/12/2023] Open
Abstract
Autophagy, a critical catabolic process for cell survival against different types of stress, has a role in the differentiation of various cells, such as cardiomyocytes. Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) is an energy-sensing protein kinase involved in the regulation of autophagy. In addition to its direct role in regulating autophagy, AMPK can also influence other cellular processes by regulating mitochondrial function, posttranslational acetylation, cardiomyocyte metabolism, mitochondrial autophagy, endoplasmic reticulum stress, and apoptosis. As AMPK is involved in the control of various cellular processes, it can influence the health and survival of cardiomyocytes. This study investigated the effects of an AMPK inducer (Metformin) and an autophagy inhibitor (Hydroxychloroquine) on the differentiation of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs). The results showed that autophagy was upregulated during cardiac differentiation. Furthermore, AMPK activation increased the expression of CM-specific markers in hPSC-CMs. Additionally, autophagy inhibition impaired cardiomyocyte differentiation by targeting autophagosome-lysosome fusion. These results indicate the significance of autophagy in cardiomyocyte differentiation. In conclusion, AMPK might be a promising target for the regulation of cardiomyocyte generation by in vitro differentiation of pluripotent stem cells.
Collapse
Affiliation(s)
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | | | - Yaser Tahamtani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Reproductive Epidemiology Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Mehdi Totonchi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran.
| |
Collapse
|
32
|
Liu X, Lin C, Zhong W, Yuan Z, Yan P, Guan S. Effective Attenuation of Arteriosclerosis Following Lymphatic-Targeted Delivery of Hyaluronic Acid-Decorated Rapamycin Liposomes. Int J Nanomedicine 2023; 18:4403-4419. [PMID: 37551276 PMCID: PMC10404413 DOI: 10.2147/ijn.s410653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/10/2023] [Indexed: 08/09/2023] Open
Abstract
Background The activation of lymphatic vessel function is the crux to resolving atherosclerosis (AS), a chronic inflammatory disease. Rapamycin (RAPA) recently has attracted considerable attention as a potent drug to induce atherosclerotic plaque attenuation. The objective of this work was to develop a ligand-decorated, RAPA-loaded liposome for lymphatic-targeted delivery of drugs to improve abnormal lymphatic structure and function, resulting in highly effective regression of atherosclerotic plaques. Methods Hyaluronic acid-decorated, RAPA-loaded liposomes (HA-RL) were fabricated by emulsion-solvent evaporation. The average size, zeta potential, entrapment efficiency were characterized, and the stability and drug release in vitro were investigated. Furthermore, the in vitro and in vivo lymphatic targeting ability were evaluated on lymphatic endothelial cells and LDLR-/- mice, and the efficiency of this nano-system in inducing the attenuation of atherosclerotic plaques was confirmed. Results HA-RL had a size of 100 nm, over 90% drug encapsulation efficiency, the storage stability was distinguished, demonstrating a slow release from the lipid nano-carriers. The mean retention time (MRT) and elimination half-life (t1/2β) achieved from HA-RL were 100.27±73.08 h and 70.74±50.80 h, respectively. HA-RL acquired the most prominent efficacy of lymphatic-targeted delivery and atherosclerotic plaques attenuation, implying the successful implementation of this novel drug delivery system in vivo. Conclusion HA-RL exhibited the most appreciable lymphatic targeting ability and best atherosclerotic plaques attenuation efficiency, opening a new paradigm and promising perspective for the treatment of arteriosclerosis.
Collapse
Affiliation(s)
- Xiaojia Liu
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| | - Caiyan Lin
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Wenfei Zhong
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Zhongwen Yuan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Pengke Yan
- Department of Pharmacy, Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Shixia Guan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, People’s Republic of China
| |
Collapse
|
33
|
Chi C, Song K. Cellular reprogramming of fibroblasts in heart regeneration. J Mol Cell Cardiol 2023; 180:84-93. [PMID: 36965699 PMCID: PMC10347886 DOI: 10.1016/j.yjmcc.2023.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/10/2023] [Accepted: 03/21/2023] [Indexed: 03/27/2023]
Abstract
Myocardial infarction causes the loss of cardiomyocytes and the formation of cardiac fibrosis due to the activation of cardiac fibroblasts, leading to cardiac dysfunction and heart failure. Unfortunately, current therapeutic interventions can only slow the disease progression. Furthermore, they cannot fully restore cardiac function, likely because the adult human heart lacks sufficient capacity to regenerate cardiomyocytes. Therefore, intensive efforts have focused on developing therapeutics to regenerate the damaged heart. Several strategies have been intensively investigated, including stimulation of cardiomyocyte proliferation, transplantation of stem cell-derived cardiomyocytes, and conversion of fibroblasts into cardiac cells. Resident cardiac fibroblasts are critical in the maintenance of the structure and contractility of the heart. Fibroblast plasticity makes this type of cells be reprogrammed into many cell types, including but not limited to induced pluripotent stem cells, induced cardiac progenitor cells, and induced cardiomyocytes. Fibroblasts have become a therapeutic target due to their critical roles in cardiac pathogenesis. This review summarizes the reprogramming of fibroblasts into induced pluripotent stem cell-derived cardiomyocytes, induced cardiac progenitor cells, and induced cardiomyocytes to repair a damaged heart, outlines recent findings in utilizing fibroblast-derived cells for heart regeneration, and discusses the limitations and challenges.
Collapse
Affiliation(s)
- Congwu Chi
- Division of Cardiology, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kunhua Song
- Division of Cardiology, Department of Medicine, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Gates Center for Regenerative Medicine and Stem Cell Biology, The University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
34
|
Korover N, Etzion S, Cherniak A, Rabinski T, Levitas A, Etzion Y, Ofir R, Parvari R, Cohen S. Functional defects in hiPSCs-derived cardiomyocytes from patients with a PLEKHM2-mutation associated with dilated cardiomyopathy and left ventricular non-compaction. Biol Res 2023; 56:34. [PMID: 37349842 PMCID: PMC10288792 DOI: 10.1186/s40659-023-00442-5] [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: 11/16/2022] [Accepted: 05/17/2023] [Indexed: 06/24/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a primary myocardial disease, leading to heart failure and excessive risk of sudden cardiac death with rather poorly understood pathophysiology. In 2015, Parvari's group identified a recessive mutation in the autophagy regulator, PLEKHM2 gene, in a family with severe recessive DCM and left ventricular non-compaction (LVNC). Fibroblasts isolated from these patients exhibited abnormal subcellular distribution of endosomes, Golgi apparatus, lysosomes and had impaired autophagy flux. To better understand the effect of mutated PLEKHM2 on cardiac tissue, we generated and characterized induced pluripotent stem cells-derived cardiomyocytes (iPSC-CMs) from two patients and a healthy control from the same family. The patient iPSC-CMs showed low expression levels of genes encoding for contractile functional proteins (α and β-myosin heavy chains and 2v and 2a-myosin light chains), structural proteins integral to heart contraction (Troponin C, T and I) and proteins participating in Ca2+ pumping action (SERCA2 and Calsequestrin 2) compared to their levels in control iPSC-derived CMs. Furthermore, the sarcomeres of the patient iPSC-CMs were less oriented and aligned compared to control cells and generated slowly beating foci with lower intracellular calcium amplitude and abnormal calcium transient kinetics, measured by IonOptix system and MuscleMotion software. Autophagy in patient's iPSC-CMs was impaired as determined from a decrease in the accumulation of autophagosomes in response to chloroquine and rapamycin treatment, compared to control iPSC-CMs. Impairment in autophagy together with the deficiency in the expression of NKX2.5, MHC, MLC, Troponins and CASQ2 genes, which are related to contraction-relaxation coupling and intracellular Ca2+ signaling, may contribute to the defective function of the patient CMs and possibly affect cell maturation and cardiac failure with time.
Collapse
Affiliation(s)
- Nataly Korover
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel.
| | - Sharon Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Alexander Cherniak
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Tatiana Rabinski
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Aviva Levitas
- Department of Pediatric Cardiology, Soroka University Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Yoram Etzion
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| | - Rivka Ofir
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Dead Sea & Arava Science Center, 8691000, Masada, Israel
| | - Ruti Parvari
- Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Smadar Cohen
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer-Sheva, Israel
| |
Collapse
|
35
|
Ruiz-Velasco A, Raja R, Chen X, Ganenthiran H, Kaur N, Alatawi NHO, Miller JM, Abouleisa RR, Ou Q, Zhao X, Fonseka O, Wang X, Hille SS, Frey N, Wang T, Mohamed TM, Müller OJ, Cartwright EJ, Liu W. Restored autophagy is protective against PAK3-induced cardiac dysfunction. iScience 2023; 26:106970. [PMID: 37324527 PMCID: PMC10265534 DOI: 10.1016/j.isci.2023.106970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/27/2023] [Accepted: 05/23/2023] [Indexed: 06/17/2023] Open
Abstract
Despite the development of clinical treatments, heart failure remains the leading cause of mortality. We observed that p21-activated kinase 3 (PAK3) was augmented in failing human and mouse hearts. Furthermore, mice with cardiac-specific PAK3 overexpression exhibited exacerbated pathological remodeling and deteriorated cardiac function. Myocardium with PAK3 overexpression displayed hypertrophic growth, excessive fibrosis, and aggravated apoptosis following isoprenaline stimulation as early as two days. Mechanistically, using cultured cardiomyocytes and human-relevant samples under distinct stimulations, we, for the first time, demonstrated that PAK3 acts as a suppressor of autophagy through hyper-activation of the mechanistic target of rapamycin complex 1 (mTORC1). Defective autophagy in the myocardium contributes to the progression of heart failure. More importantly, PAK3-provoked cardiac dysfunction was mitigated by administering an autophagic inducer. Our study illustrates a unique role of PAK3 in autophagy regulation and the therapeutic potential of targeting this axis for heart failure.
Collapse
Affiliation(s)
- Andrea Ruiz-Velasco
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Rida Raja
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Xinyi Chen
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Haresh Ganenthiran
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Namrita Kaur
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Nasser hawimel o Alatawi
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Jessica M. Miller
- Institute of Molecular Cardiology, University of Louisville, 580 S Preston St, Louisville, KY 40202, USA
| | - Riham R.E. Abouleisa
- Institute of Molecular Cardiology, University of Louisville, 580 S Preston St, Louisville, KY 40202, USA
| | - Qinghui Ou
- Institute of Molecular Cardiology, University of Louisville, 580 S Preston St, Louisville, KY 40202, USA
| | - Xiangjun Zhao
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Oveena Fonseka
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Xin Wang
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Susanne S. Hille
- Department of Internal Medicine III, University of Kiel, Kiel, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Norbert Frey
- Department of Internal Medicine III, University of Kiel, Kiel, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Tao Wang
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Tamer M.A. Mohamed
- Institute of Molecular Cardiology, University of Louisville, 580 S Preston St, Louisville, KY 40202, USA
| | - Oliver J. Müller
- Department of Internal Medicine III, University of Kiel, Kiel, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Hamburg/Kiel/Lübeck, Kiel, Germany
| | - Elizabeth J. Cartwright
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Wei Liu
- Faculty of Biology, Medicine, and Health, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
36
|
Zou L, Zhang Y, Cheraga N, Abodunrin OD, Qu KY, Qiao L, Ma YQ, Chen LJ, Huang NP. Chlorin e6 (Ce6)-loaded plaque-specific liposome with enhanced photodynamic therapy effect for atherosclerosis treatment. Talanta 2023; 265:124772. [PMID: 37327664 DOI: 10.1016/j.talanta.2023.124772] [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: 03/02/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/18/2023]
Abstract
Recently, photodynamic therapy (PDT) has been considered as a new strategy for atherosclerosis treatment. Targeted delivery of photosensitizer could significantly reduce its toxicity and enhance its phototherapeutic efficiency. CD68 is an antibody that can be conjugated to nano-drug delivery systems to actively target plaque sites, owing to its specific binding to CD68 receptors that are highly expressed on the surfaces of macrophage-derived foam cells. Liposomes are very popular nanocarriers due to their ability to encapsulate a wide range of therapeutic compounds including drugs, microRNAs and photosensitizers, and their ability to be surface-modified with targeting moieties leading to the development of nanocarriers with an improved targeted ability. Hence, we designed a Ce6-loaded liposomes using the film dispersion method, followed by the conjugation of CD68 antibody on the liposomal surface through a covalent crosslinking reaction, forming CD68-modified Ce6-loaded liposomes (CD68-Ce6-mediated liposomes). Flow cytometry results indicated that Ce6-containing liposomes were more effective in promoting intracellular uptake after laser irradiation. Furthermore, CD68-modified liposomes significantly strengthened the cellular recognization and thus internalization. Different cell lines have been incubated with the liposomes, and the results showed that CD68-Ce6-mediated liposomes had no significant cytotoxicity to coronary artery endothelial cells (HCAEC) under selected conditions. Interestingly, they promoted autophagy in foam cells through the increase in LC3-Ⅰ, LC3-Ⅱ expression and the decrease in p62 expression, and restrained the migration of mouse aortic vascular smooth muscle cells (MOVAS) in vitro. Moreover, the enhancement of atherosclerotic plaque stability and the reduction in the cholesterol content by CD68-Ce6-mediated liposomes were dependent on transient reactive oxygen species (ROS) generated under laser irradiation. In summary, we demonstrated that CD68-Ce6-mediated liposomes, as a photosensitizer nano-drug delivery system, have an inhibitory effect on MOVAS migration and a promotion of cholesterol efflux in foam cells, and thereby, represent promising nanocarriers for atherosclerosis photodynamic therapy.
Collapse
Affiliation(s)
- Lin Zou
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yao Zhang
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China
| | - Nihad Cheraga
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Oluwatosin David Abodunrin
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Kai-Yun Qu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Li Qiao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yu-Qing Ma
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Li-Juan Chen
- Department of Cardiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, China; Department of Cardiology, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Nanjing, 211200, China.
| | - Ning-Ping Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
37
|
Juni RP, Kocken JMM, Abreu RC, Ottaviani L, Davalan T, Duygu B, Poels EM, Vasilevich A, Hegenbarth JC, Appari M, Bitsch N, Olieslagers S, Schrijvers DM, Stoll M, Heineke J, de Boer J, de Windt LJ, da Costa Martins PA. MicroRNA-216a is essential for cardiac angiogenesis. Mol Ther 2023; 31:1807-1828. [PMID: 37073128 PMCID: PMC10277893 DOI: 10.1016/j.ymthe.2023.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/18/2023] [Accepted: 04/12/2023] [Indexed: 04/20/2023] Open
Abstract
While it is experimentally supported that impaired myocardial vascularization contributes to a mismatch between myocardial oxygen demand and supply, a mechanistic basis for disruption of coordinated tissue growth and angiogenesis in heart failure remains poorly understood. Silencing strategies that impair microRNA biogenesis have firmly implicated microRNAs in the regulation of angiogenesis, and individual microRNAs prove to be crucial in developmental or tumor angiogenesis. A high-throughput functional screening for the analysis of a whole-genome microRNA silencing library with regard to their phenotypic effect on endothelial cell proliferation as a key parameter, revealed several anti- and pro-proliferative microRNAs. Among those was miR-216a, a pro-angiogenic microRNA which is enriched in cardiac microvascular endothelial cells and reduced in expression under cardiac stress conditions. miR-216a null mice display dramatic cardiac phenotypes related to impaired myocardial vascularization and unbalanced autophagy and inflammation, supporting a model where microRNA regulation of microvascularization impacts the cardiac response to stress.
Collapse
Affiliation(s)
- Rio P Juni
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Physiology, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Jordy M M Kocken
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Ricardo C Abreu
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands; Biomaterials and Stem Cell Based Therapeutics Group, CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Coimbra, Portugal
| | - Lara Ottaviani
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Tim Davalan
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Burcu Duygu
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Ella M Poels
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Aliaksei Vasilevich
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, University of Eindhoven, Eindhoven, the Netherlands
| | - Jana C Hegenbarth
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Mahesh Appari
- Division of Cardiovascular Medicine, British Heart Foundation Centre of Research Excellence, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU United Kingdom
| | - Nicole Bitsch
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Serve Olieslagers
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Dorien M Schrijvers
- Laboratory of Physiopharmacology, University of Antwerp, 2610 Wilrijk, Belgium
| | - Monika Stoll
- Department of Genetic Epidemiology, Institute of Human Genetics, University of Münster, 48149 Münster, Germany; Department of Biochemistry, CARIM School for Cardiovascular Diseases, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Joerg Heineke
- Department of Cardiovascular Physiology, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; DZHK, Partner Site Heidelberg/Mannheim, 69120 Heidelberg, Germany
| | - Jan de Boer
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, University of Eindhoven, Eindhoven, the Netherlands
| | - Leon J de Windt
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands
| | - Paula A da Costa Martins
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 ER Maastricht, the Netherlands; Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal.
| |
Collapse
|
38
|
Ouyang X, Bakshi S, Benavides GA, Sun Z, Hernandez‐Moreno G, Collins HE, Kane MS, Litovsky S, Young ME, Chatham JC, Darley‐Usmar V, Wende AR, Zhang J. Cardiomyocyte ZKSCAN3 regulates remodeling following pressure-overload. Physiol Rep 2023; 11:e15686. [PMID: 37144628 PMCID: PMC10161215 DOI: 10.14814/phy2.15686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 05/06/2023] Open
Abstract
Autophagy is important for protein and organelle quality control. Growing evidence demonstrates that autophagy is tightly controlled by transcriptional mechanisms, including repression by zinc finger containing KRAB and SCAN domains 3 (ZKSCAN3). We hypothesize that cardiomyocyte-specific ZKSCAN3 knockout (Z3K) disrupts autophagy activation and repression balance and exacerbates cardiac pressure-overload-induced remodeling following transverse aortic constriction (TAC). Indeed, Z3K mice had an enhanced mortality compared to control (Con) mice following TAC. Z3K-TAC mice that survived exhibited a lower body weight compared to Z3K-Sham. Although both Con and Z3K mice exhibited cardiac hypertrophy after TAC, Z3K mice exhibited TAC-induced increase of left ventricular posterior wall thickness at end diastole (LVPWd). Conversely, Con-TAC mice exhibited decreases in PWT%, fractional shortening (FS%), and ejection fraction (EF%). Autophagy genes (Tfeb, Lc3b, and Ctsd) were decreased by the loss of ZKSCAN3. TAC suppressed Zkscan3, Tfeb, Lc3b, and Ctsd in Con mice, but not in Z3K. The Myh6/Myh7 ratio, which is related to cardiac remodeling, was decreased by the loss of ZKSCAN3. Although Ppargc1a mRNA and citrate synthase activities were decreased by TAC in both genotypes, mitochondrial electron transport chain activity did not change. Bi-variant analyses show that while in Con-Sham, the levels of autophagy and cardiac remodeling mRNAs form a strong correlation network, such was disrupted in Con-TAC, Z3K-Sham, and Z3K-TAC. Ppargc1a also forms different links in Con-sham, Con-TAC, Z3K-Sham, and Z3K-TAC. We conclude that ZKSCAN3 in cardiomyocytes reprograms autophagy and cardiac remodeling gene transcription, and their relationships with mitochondrial activities in response to TAC-induced pressure overload.
Collapse
Affiliation(s)
- Xiaosen Ouyang
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Sayan Bakshi
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Gloria A. Benavides
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Zhihuan Sun
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Gerardo Hernandez‐Moreno
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Present address:
Department of Materials Science and Engineering, Laboratory for Polymers & Healthcare Materials/DevicesThe University of Alabama at Birmingham (UAB)BirminghamALUSA
| | - Helen E. Collins
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Present address:
Division of Environmental Medicine, Center for Cardiometabolic ScienceThe University of LouisvilleLouisvilleKYUSA
| | - Mariame S. Kane
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Present address:
Birmingham VA Health Care System (BVACS)ALUSA
| | - Silvio Litovsky
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Martin E. Young
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - John C. Chatham
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Victor Darley‐Usmar
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Adam R. Wende
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Jianhua Zhang
- Department of PathologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Birmingham VA Medical CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
39
|
Bonilla IM, Baine S, Pokrass A, Mariángelo JIE, Kalyanasundaram A, Bogdanov V, Mezache L, Sakuta G, Beard CM, Belevych A, Tikunova S, Terentyeva R, Terentyev D, Davis J, Veeraraghavan R, Carnes CA, Györke S. STIM1 ablation impairs exercise-induced physiological cardiac hypertrophy and dysregulates autophagy in mouse hearts. J Appl Physiol (1985) 2023; 134:1287-1299. [PMID: 36995910 PMCID: PMC10190841 DOI: 10.1152/japplphysiol.00363.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 03/13/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023] Open
Abstract
Cardiac stromal interaction molecule 1 (STIM1), a key mediator of store-operated Ca2+ entry (SOCE), is a known determinant of cardiomyocyte pathological growth in hypertrophic cardiomyopathy. We examined the role of STIM1 and SOCE in response to exercise-dependent physiological hypertrophy. Wild-type (WT) mice subjected to exercise training (WT-Ex) showed a significant increase in exercise capacity and heart weight compared with sedentary (WT-Sed) mice. Moreover, myocytes from WT-Ex hearts displayed an increase in length, but not width, compared with WT-Sed myocytes. Conversely, exercised cardiac-specific STIM1 knock-out mice (cSTIM1KO-Ex), although displaying significant increase in heart weight and cardiac dilation, evidenced no changes in myocyte size and displayed a decreased exercise capacity, impaired cardiac function, and premature death compared with sedentary cardiac-specific STIM1 knock-out mice (cSTIM1KO-Sed). Confocal Ca2+ imaging demonstrated enhanced SOCE in WT-Ex myocytes compared with WT-Sed myocytes with no measurable SOCE detected in cSTIM1KO myocytes. Exercise training induced a significant increase in cardiac phospho-Akt Ser473 in WT mice but not in cSTIM1KO mice. No differences were observed in phosphorylation of mammalian target of rapamycin (mTOR) and glycogen synthase kinase (GSK) in exercised versus sedentary cSTIM1KO mice hearts. cSTIM1KO-Sed mice showed increased basal MAPK phosphorylation compared with WT-Sed that was not altered by exercise training. Finally, histological analysis revealed exercise resulted in increased autophagy in cSTIM1KO but not in WT myocytes. Taken together, our results suggest that adaptive cardiac hypertrophy in response to exercise training involves STIM1-mediated SOCE. Our results demonstrate that STIM1 is involved in and essential for the myocyte longitudinal growth and mTOR activation in response to endurance exercise training.NEW & NOTEWORTHY Store-operated Ca2+ entry (SOCE) has been implicated in pathological cardiac hypertrophy; however, its role in physiological hypertrophy is unknown. Here we report that SOCE is also essential for physiological cardiac hypertrophy and functional adaptations in response to endurance exercise. These adaptations were associated with activation of AKT/mTOR pathway and curtailed cardiac autophagy and degeneration. Thus, SOCE is a common mechanism and an important bifurcation point for signaling paths involved in physiological and pathological hypertrophy.
Collapse
Affiliation(s)
- Ingrid M Bonilla
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Veterans Affairs Caribbean Healthcare System, San Juan, Puerto Rico, United States
- Department of Pharmacology and Toxicology, School of Medicine, University of Puerto Rico, San Juan, Puerto Rico, United States
| | - Stephen Baine
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Anastasia Pokrass
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Juan Ignacio Elio Mariángelo
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Anuradha Kalyanasundaram
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
- Bob and Corrine Frick Center for Heart Failure and Arrhythmia, Dorothy M. Davis Heart & Lung Research Institute, Columbus, Ohio, United States
| | - Vladimir Bogdanov
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Louisa Mezache
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Galina Sakuta
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Casey M Beard
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Andriy Belevych
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Svetlana Tikunova
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Radmila Terentyeva
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Dmitry Terentyev
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Jonathan Davis
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Rengasayee Veeraraghavan
- Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus, Ohio, United States
| | - Cynthia A Carnes
- Department of Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, United States
| | - Sandor Györke
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
40
|
Cho JM, Park SK, Kwon OS, Taylor La Salle D, Cerbie J, Fermoyle CC, Morgan D, Nelson A, Bledsoe A, Bharath LP, Tandar M, Kunapuli SP, Richardson RS, Anandh Babu PV, Mookherjee S, Kishore BK, Wang F, Yang T, Boudina S, Trinity JD, Symons JD. Activating P2Y1 receptors improves function in arteries with repressed autophagy. Cardiovasc Res 2023; 119:252-267. [PMID: 35420120 PMCID: PMC10236004 DOI: 10.1093/cvr/cvac061] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 02/19/2022] [Accepted: 03/23/2022] [Indexed: 11/14/2022] Open
Abstract
AIM The importance of endothelial cell (EC) autophagy to vascular homeostasis in the context of health and disease is evolving. Earlier, we reported that intact EC autophagy is requisite to maintain shear-stress-induced nitric oxide (NO) generation via glycolysis-dependent purinergic signalling to endothelial NO synthase (eNOS). Here, we illustrate the translational and functional significance of these findings. METHODS AND RESULTS First, we assessed translational relevance using older male humans and mice that exhibit blunted EC autophagy and impaired arterial function vs. adult controls. Active hyperaemia evoked by rhythmic handgrip exercise-elevated radial artery shear-rate similarly from baseline in adult and older subjects for 60 min. Compared with baseline, indexes of autophagy initiation, p-eNOSS1177 activation, and NO generation, occurred in radial artery ECs obtained from adult but not older volunteers. Regarding mice, indexes of autophagy and p-eNOSS1177 activation were robust in ECs from adult but not older animals that completed 60-min treadmill-running. Furthermore, 20 dyne • cm2 laminar shear stress × 45-min increased autophagic flux, glycolysis, ATP production, and p-eNOSS1177 in primary arterial ECs obtained from adult but not older mice. Concerning functional relevance, we next questioned whether the inability to initiate EC autophagy, glycolysis, and p-eNOSS1177in vitro precipitates arterial dysfunction ex vivo. Compromised intraluminal flow-mediated vasodilation displayed by arteries from older vs. adult mice was recapitulated in vessels from adult mice by (i) NO synthase inhibition; (ii) acute autophagy impairment using 3-methyladenine (3-MA); (iii) EC Atg3 depletion (iecAtg3KO mice); (iv) purinergic 2Y1-receptor (P2Y1-R) blockade; and (v) germline depletion of P2Y1-Rs. Importantly, P2Y1-R activation using 2-methylthio-ADP (2-Me-ADP) improved vasodilatory capacity in arteries from (i) adult mice treated with 3-MA; (ii) adult iecAtg3KO mice; and (iii) older animals with repressed EC autophagy. CONCLUSIONS Arterial dysfunction concurrent with pharmacological, genetic, and age-associated EC autophagy compromise is improved by activating P2Y1-Rs.
Collapse
Affiliation(s)
- Jae Min Cho
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Seul-Ki Park
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, CT, USA
- Department of Orthopedic Surgery & Center on Aging, University of Connecticut School of Medicine, Storrs, CT, USA
| | - David Taylor La Salle
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - James Cerbie
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Caitlin C Fermoyle
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - David Morgan
- Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Ashley Nelson
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
| | - Amber Bledsoe
- Department of Anesthesiology, University of Utah, Salt Lake City, UT, USA
| | - Leena P Bharath
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Megan Tandar
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Satya P Kunapuli
- Sol Sherry Thrombosis Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Russell S Richardson
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | | | - Sohom Mookherjee
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Bellamkonda K Kishore
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Nephrology Research, George E. Whalen VA Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Nephrology, University of Utah, Salt Lake City, UT, USA
| | - Fei Wang
- Nephrology Research, George E. Whalen VA Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Nephrology, University of Utah, Salt Lake City, UT, USA
| | - Tianxin Yang
- Nephrology Research, George E. Whalen VA Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Nephrology, University of Utah, Salt Lake City, UT, USA
| | - Sihem Boudina
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Joel D Trinity
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans Affairs Medical Center, Salt Lake City, UT, USA
- Department of Internal Medicine, Division of Geriatrics, University of Utah, Salt Lake City, UT, USA
| | - John David Symons
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
- Division of Endocrinology, Metabolism and Diabetes, Program in Molecular Medicine University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
41
|
Biochemical Mechanisms beyond Glycosphingolipid Accumulation in Fabry Disease: Might They Provide Additional Therapeutic Treatments? J Clin Med 2023; 12:jcm12052063. [PMID: 36902850 PMCID: PMC10004377 DOI: 10.3390/jcm12052063] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Fabry disease is a rare X-linked disease characterized by deficient expression and activity of alpha-galactosidase A (α-GalA) with consequent lysosomal accumulation of glycosphingolipid in various organs. Currently, enzyme replacement therapy is the cornerstone of the treatment of all Fabry patients, although in the long-term it fails to completely halt the disease's progression. This suggests on one hand that the adverse outcomes cannot be justified only by the lysosomal accumulation of glycosphingolipids and on the other that additional therapies targeted at specific secondary mechanisms might contribute to halt the progression of cardiac, cerebrovascular, and renal disease that occur in Fabry patients. Several studies reported how secondary biochemical processes beyond Gb3 and lyso-Gb3 accumulation-such as oxidative stress, compromised energy metabolism, altered membrane lipid, disturbed cellular trafficking, and impaired autophagy-might exacerbate Fabry disease adverse outcomes. This review aims to summarize the current knowledge of these pathogenetic intracellular mechanisms in Fabry disease, which might suggest novel additional strategies for its treatment.
Collapse
|
42
|
Chronic intermittent hypoxia-induced BNIP3 expression mitigates contractile dysfunction and myocardial injury in animal and cell model via modulating autophagy. Hum Cell 2023; 36:631-642. [PMID: 36627546 DOI: 10.1007/s13577-022-00851-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 12/20/2022] [Indexed: 01/11/2023]
Abstract
Obstructive sleep apnea syndrome is generally associated with multiple cardiovascular disorders, such as myocardial hypertrophy. Autophagy is strictly modulated to maintain cardiac homeostasis. Post-injury autophagy is closely associated with pathological cardiac hypertrophy. BCL2 interacting protein 3 (BNIP3) and BNIP3-like protein (BNIP3L) can cause cell death and are important for hypoxia-elicited autophagy. Here, we evaluated whether BNIP3 could mitigate functional remodeling and cardiac hypertrophy through regulation of autophagy. Male WT rats or rats with BNIP3 knockout were subjected to chronic intermittent hypoxia (CIH) for 8 h/day over 5 weeks. Echocardiography and morphology were employed to assess the cardioprotective effects. Autophagy was assessed via transmission electron microscopy and detection of LC3, p62, and Beclin-1. Terminal deoxynucleotidyl transferase dUTP nick end labeling and the Bax/Bcl2 ratio were used to monitor apoptosis. Biochemical evaluations were performed to assess oxidative stress. Additionally, BNIP3-knockdown H9c2 cells that were subjected to CIH were used to examine autophagy and apoptosis to confirm the findings of the animal study. The CIH group showed elevated heart weight/body weight and left ventricle weight/body weight proportions, along with left ventricular remodeling. CIH-exposed rats exhibited dramatically higher fractional shortening and ejection fractions than the controls. In addition, the levels of autophagy markers Beclin-1 and LC3-II/I were increased, whereas the level of p62 was reduced by CIH treatment. The oxidative marker levels and the apoptosis index in the CIH group were markedly increased. Knockout of BNIP3 significantly aggravated the impairment in cardiac function, apoptosis, oxidative stress, and hypertrophy of CIH rats, while significantly reducing autophagy. The autophagy-associated PI3K/Akt/mTOR pathway was also deactivated by BNIP3 knockout. At the cellular level, CIH treatment significantly upregulated autophagy and apoptosis; however, BNIP3 silencing reduced autophagy and promoted apoptosis. CIH treatment-mediated upregulation of BNIP3 expression plays a crucial role in autophagy by targeting the PI3K/Akt/mTOR pathway, alleviating cardiac hypertrophy.
Collapse
|
43
|
Madder ( Rubia cordifolia L.) Alleviates Myocardial Ischemia-Reperfusion Injury by Protecting Endothelial Cells from Apoptosis and Inflammation. Mediators Inflamm 2023; 2023:5015039. [PMID: 36875688 PMCID: PMC9981279 DOI: 10.1155/2023/5015039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 02/25/2023] Open
Abstract
Objective Ischemia-reperfusion injury often occurs in organ transplantation, coronary heart disease, ischemic heart disease, and other diseases, which greatly reduces clinical efficacy. This study examined the effectiveness of madder as a medicine to treat ischemia-reperfusion injury. Methods The efficacy of madder was evaluated by measuring myocardial infarction size, coronary outflow volume, myocardial contraction rate, activation of inflammatory factors, autophagy factors, apoptosis factors, and related pathway genes in mice. Results The results indicated that treatment with madder can effectively reduce the area of myocardial infarction and restore arterial blood flow velocity and myocardial contractility in mice. Additionally, madder treatment inhibited the expression of inflammatory factors, autophagy factors, and apoptosis factors in mice and reduced the degree of myocardial cell injury. Studies have also shown that madder treatment can alleviate myocardial ischemia-reperfusion injury in mice and inhibit the occurrence of inflammatory response by inhibiting the activity of the NF-κB pathway. Conclusion The results showed that madder was effective against ischemia-reperfusion injury, thus showing potential as a clinical drug for treating ischemia-reperfusion injury.
Collapse
|
44
|
Feng S, Huang F, Zhang Y, Feng Y, Zhang Y, Cao Y, Wang X. The pathophysiological and molecular mechanisms of atmospheric PM 2.5 affecting cardiovascular health: A review. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 249:114444. [PMID: 38321663 DOI: 10.1016/j.ecoenv.2022.114444] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 02/08/2024]
Abstract
BACKGROUND Exposure to ambient fine particulate matter (PM2.5, with aerodynamic diameter less than 2.5 µm) is a leading environmental risk factor for global cardiovascular health concern. OBJECTIVE To provide a roadmap for those new to this field, we reviewed the new insights into the pathophysiological and cellular/molecular mechanisms of PM2.5 responsible for cardiovascular health. MAIN FINDINGS PM2.5 is able to disrupt multiple physiological barriers integrity and translocate into the systemic circulation and get access to a range of secondary target organs. An ever-growing body of epidemiological and controlled exposure studies has evidenced a causal relationship between PM2.5 exposure and cardiovascular morbidity and mortality. A variety of cellular and molecular biology mechanisms responsible for the detrimental cardiovascular outcomes attributable to PM2.5 exposure have been described, including metabolic activation, oxidative stress, genotoxicity, inflammation, dysregulation of Ca2+ signaling, disturbance of autophagy, and induction of apoptosis, by which PM2.5 exposure impacts the functions and fates of multiple target cells in cardiovascular system or related organs and further alters a series of pathophysiological processes, such as cardiac autonomic nervous system imbalance, increasing blood pressure, metabolic disorder, accelerated atherosclerosis and plaque vulnerability, platelet aggregation and thrombosis, and disruption in cardiac structure and function, ultimately leading to cardiovascular events and death. Therein, oxidative stress and inflammation were suggested to play pivotal roles in those pathophysiological processes. CONCLUSION Those biology mechanisms have deepen insights into the etiology, course, prevention and treatment of this public health concern, although the underlying mechanisms have not yet been entirely clarified.
Collapse
Affiliation(s)
- Shaolong Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China; Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China; The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China.
| | - Fangfang Huang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yuqi Zhang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yashi Feng
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Ying Zhang
- The Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Heath, School of Public Health, Guilin Medical University, Guilin 541199, China
| | - Yunchang Cao
- The Department of Molecular Biology, School of Intelligent Medicine and Biotechnology, Guilin Medical University, Guilin 541199, China
| | - Xinming Wang
- Guangdong Provincial Key Laboratory of Environmental Protection and Resources Utilization, Guangzhou 510640, China; The State Key Laboratory of Organic Geochemistry, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou 510640, China
| |
Collapse
|
45
|
Titin-truncating variants in hiPSC cardiomyocytes induce pathogenic proteinopathy and sarcomere defects with preserved core contractile machinery. Stem Cell Reports 2022; 18:220-236. [PMID: 36525964 PMCID: PMC9860080 DOI: 10.1016/j.stemcr.2022.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 12/23/2022] Open
Abstract
Titin-truncating variants (TTNtv) are the single largest genetic cause of dilated cardiomyopathy (DCM). In this study we modeled disease phenotypes of A-band TTNtv-induced DCM in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) using genome editing and tissue engineering technologies. Transcriptomic, cellular, and micro-tissue studies revealed that A-band TTNtv hiPSC-CMs exhibit pathogenic proteinopathy, sarcomere defects, aberrant Na+ channel activities, and contractile dysfunction. These phenotypes establish a dual mechanism of poison peptide effect and haploinsufficiency that collectively contribute to DCM pathogenesis. However, TTNtv cellular defects did not interfere with the function of the core contractile machinery, the actin-myosin-troponin-Ca2+ complex, and preserved the therapeutic mechanism of sarcomere modulators. Treatment of TTNtv cardiac micro-tissues with investigational sarcomere modulators augmented contractility and resulted in sustained transcriptomic changes that promote reversal of DCM disease signatures. Together, our findings elucidate the underlying pathogenic mechanisms of A-band TTNtv-induced DCM and demonstrate the validity of sarcomere modulators as potential therapeutics.
Collapse
|
46
|
Zeng Y, Ren WQ, Wen AZ, Zhang W, Fan FY, Chen OY. Autophagy and pressure overload-induced cardiac hypertrophy. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2022; 24:1101-1108. [PMID: 35043747 DOI: 10.1080/10286020.2021.2024810] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
Autophagy plays an important role in the pathogenesis of cardiovascular diseases. Dysregulation of autophagy may have a huge effect on cardiac hypertrophy induced by overload pressure although reports on autophagy and cardiac hypertrophy have been contradictory. Some studies showed that autophagy activation attenuated cardiac hypertrophy. However, others suggested that inhibition of autophagy would be protective. Different research models or different pathways involved could be responsible for it. Cardiac hypertrophy may be alleviated through regulation of autophagy. This review aims to highlight the pathways and therapeutic targets identified in the prevention and treatment of cardiac hypertrophy by regulating autophagy.
Collapse
Affiliation(s)
- Yong Zeng
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Wei-Qiong Ren
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Ai-Zhen Wen
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Wen Zhang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Fu-Yuan Fan
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Ou-Ying Chen
- School of Nursing, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
47
|
Mokhtari B, Badalzadeh R. Protective and deleterious effects of autophagy in the setting of myocardial ischemia/reperfusion injury: an overview. Mol Biol Rep 2022; 49:11081-11099. [DOI: 10.1007/s11033-022-07837-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/01/2022] [Indexed: 11/30/2022]
|
48
|
Lorusso B, Cerasoli G, Falco A, Frati C, Graiani G, Madeddu D, Nogara A, Corradini E, Roti G, Cerretani E, Gherli A, Caputi M, Gnetti L, Pilato FP, Quaini F, Lagrasta C. Β-blockers activate autophagy on infantile hemangioma-derived endothelial cells in vitro. Vascul Pharmacol 2022; 146:107110. [PMID: 36103993 DOI: 10.1016/j.vph.2022.107110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 01/18/2023]
Abstract
The mechanisms underlying the success of propranolol in the treatment of infantile hemangioma (IH) remain elusive and do not fully explain the rapid regression of hemangiomatous lesions following drug administration. As autophagy is critically implicated in vascular homeostasis, we determined whether β-blockers trigger the autophagic flux on infantile hemangioma-derived endothelial cells (Hem-ECs) in vitro. MATERIAL AND METHODS Fresh tissue specimens, surgically removed for therapeutic purpose to seven children affected by proliferative IH, were subjected to enzymatic digestion. Cells were sorted with anti-human CD31 immunolabeled magnetic microbeads. Following phenotypic characterization, expanded Hem-ECs, at P2 to P6, were exposed to different concentrations (50 μM to 150 μM) of propranolol, atenolol or metoprolol alone and in combination with the autophagy inhibitor Bafilomycin A1. Rapamycin, a potent inducer of autophagy, was also used as control. Autophagy was assessed by Lysotracker Red staining, western blot analysis of LC3BII/LC3BI and p62, and morphologically by transmission electron microscopy. RESULTS Hem-ECs treated with either propranolol, atenolol or metoprolol displayed positive LysoTracker Red staining. Increased LC3BII/LC3BI ratio, as well as p62 modulation, were documented in β-blockers treated Hem-ECs. Abundant autophagic vacuoles and multilamellar bodies characterized the cytoplasmic ultrastructural features of autophagy in cultured Hem-ECs exposed in vitro to β-blocking agents. Importantly, similar biochemical and morphologic evidence of autophagy were observed following rapamycin while Bafilomycin A1 significantly prevented the autophagic flux promoted by β-blockers in Hem-ECs. CONCLUSION Our data suggest that autophagy may be ascribed among the mechanisms of action of β-blockers suggesting new mechanistic insights on the potential therapeutic application of this class of drugs in pathologic conditions involving uncontrolled angiogenesis.
Collapse
Affiliation(s)
- Bruno Lorusso
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giuseppe Cerasoli
- Pediatric Surgery, Ospedale dei Bambini of Parma, University Hospital of Parma, Parma, Italy
| | - Angela Falco
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Caterina Frati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gallia Graiani
- Dental School, University of Parma Medical School, Parma, Italy
| | - Denise Madeddu
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Antonella Nogara
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Emilia Corradini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Elisa Cerretani
- Department of Medicine and Surgery, University of Parma, Parma, Italy; Department of Medical Science, University of Ferrara, Ferrara, Italy
| | - Andrea Gherli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Letizia Gnetti
- Pathology Section, University Hospital of Parma, Parma, Italy
| | | | - Federico Quaini
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Costanza Lagrasta
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| |
Collapse
|
49
|
Lu G, Wang Y, Shi Y, Zhang Z, Huang C, He W, Wang C, Shen H. Autophagy in health and disease: From molecular mechanisms to therapeutic target. MedComm (Beijing) 2022; 3:e150. [PMID: 35845350 PMCID: PMC9271889 DOI: 10.1002/mco2.150] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
Macroautophagy/autophagy is an evolutionally conserved catabolic process in which cytosolic contents, such as aggregated proteins, dysfunctional organelle, or invading pathogens, are sequestered by the double-membrane structure termed autophagosome and delivered to lysosome for degradation. Over the past two decades, autophagy has been extensively studied, from the molecular mechanisms, biological functions, implications in various human diseases, to development of autophagy-related therapeutics. This review will focus on the latest development of autophagy research, covering molecular mechanisms in control of autophagosome biogenesis and autophagosome-lysosome fusion, and the upstream regulatory pathways including the AMPK and MTORC1 pathways. We will also provide a systematic discussion on the implication of autophagy in various human diseases, including cancer, neurodegenerative disorders (Alzheimer disease, Parkinson disease, Huntington's disease, and Amyotrophic lateral sclerosis), metabolic diseases (obesity and diabetes), viral infection especially SARS-Cov-2 and COVID-19, cardiovascular diseases (cardiac ischemia/reperfusion and cardiomyopathy), and aging. Finally, we will also summarize the development of pharmacological agents that have therapeutic potential for clinical applications via targeting the autophagy pathway. It is believed that decades of hard work on autophagy research is eventually to bring real and tangible benefits for improvement of human health and control of human diseases.
Collapse
Affiliation(s)
- Guang Lu
- Department of Physiology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yu Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Yin Shi
- Department of BiochemistryZhejiang University School of MedicineHangzhouChina
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic MedicineSichuan University and Collaborative Innovation Center for BiotherapyChengduChina
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn ResearchSouthwest HospitalArmy Medical UniversityChongqingChina
| | - Chuang Wang
- Department of Pharmacology, Provincial Key Laboratory of PathophysiologyNingbo University School of MedicineNingboZhejiangChina
| | - Han‐Ming Shen
- Department of Biomedical Sciences, Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision OncologyUniversity of MacauMacauChina
| |
Collapse
|
50
|
Tian X, Xu F, Zhu Q, Feng Z, Dai W, Zhou Y, You QD, Xu X. Medicinal chemistry perspective on cGAS-STING signaling pathway with small molecule inhibitors. Eur J Med Chem 2022; 244:114791. [DOI: 10.1016/j.ejmech.2022.114791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 11/04/2022]
|