1
|
Li L, Xie S, Deng W. RNA binding proteins: Mechanistic considerations and perspectives in controlling cardiovascular diseases. Eur J Pharmacol 2025; 987:177101. [PMID: 39613174 DOI: 10.1016/j.ejphar.2024.177101] [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: 07/14/2024] [Revised: 11/01/2024] [Accepted: 11/05/2024] [Indexed: 12/01/2024]
Abstract
Cardiovascular diseases (CVDs) are becoming serious disease that endangering human health due to the increasing morbidity and mortality, and many molecular targets are involved in this complex pathologic process. Recently, RNA-binding proteins (RBPs) have received potential attention as a promising targets for preventing CVDs, including myocardial hypertrophy, dilated cardiomyopathy (DCM), myocardial fibrosis, and pulmonary hypertension (PH). As important regulators of RNA metabolism, RBPs play important roles in all steps of the gene expression cascade,and affect the occurrence and development of various diseases. In this review, we discuss the regulatory role of RBPs on various CVDs at the post transcriptional modification level based on current research. We also highlight the existing and potential RNA-based therapeutics that could impact future CVD treatments.
Collapse
Affiliation(s)
- Lanlan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
| | - Saiyang Xie
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, China; Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, China; Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
2
|
Cao Y, Yang Y, Guo C, Zong J, Li M, Li X, Yu T. Role of RNA-binding Proteins in Regulating Cell Adhesion and Progression of the Atherosclerotic Plaque and Plaque Erosion. Curr Atheroscler Rep 2024; 27:8. [PMID: 39576410 DOI: 10.1007/s11883-024-01250-2] [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] [Accepted: 10/15/2024] [Indexed: 11/24/2024]
Abstract
PURPOSE OF REVIEW RNA-binding proteins (RBPs) have emerged as crucial regulators of post-transcriptional processes, influencing the fate of RNA. This review delves into the biological functions of RBPs and their role in alternative splicing concerning atherosclerosis (AS), highlighting their participation in essential cellular processes. Our goal is to offer new insights for cardiovascular disease research and treatment. RECENT FINDING Dysregulation of RBPs is associated with various human diseases, including autoimmune and neurological disorders. The role of RBPs in the pathogenesis of AS is progressively being elucidated, as they influence plaque formation and disease progression by regulating cell function and gene expression. RBPs play intricate biological roles in regulating pre-mRNA, including editing, splicing, stability and translation. Alternative splicing has been demonstrated to enhance biological complexity and diversity. Our findings indicate that alternative splicing is extensively involved in the pathogenesis of AS. The dysregulated expression of specific RBPs in AS is linked to the production of adhesion molecules and vascular endothelium damage. Further research on RBPs could pave the way for the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Ying Cao
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, No. 308 Ningxia Road, Qingdao, 266000, People's Republic of China
| | - Chuan Guo
- Industrial Synergy Innovation Center, Linyi Vocational University of Science and Technology, Linyi, 276000, People's Republic of China
| | - Jinbao Zong
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Min Li
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China
| | - Xiaolu Li
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China
| | - Tao Yu
- Clinical Laboratory, Central Laboratory, Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, 266000, People's Republic of China.
- Department of Cardiac Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, People's Republic of China.
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, No. 38 Dengzhou Road, Qingdao, 266021, People's Republic of China.
| |
Collapse
|
3
|
Pan Z, Lv J, Zhao L, Xing K, Ye R, Zhang Y, Chen S, Yang P, Yu H, Lin Y, Li R, Wang D, Fang J, Dong Y, Sheng J, Wang X, Shan G, Zhang S, Cheng H, Xu Q, Guo X. CircARCN1 aggravates atherosclerosis by regulating HuR-mediated USP31 mRNA in macrophages. Cardiovasc Res 2024; 120:1531-1549. [PMID: 39028686 DOI: 10.1093/cvr/cvae148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/08/2024] [Accepted: 05/05/2024] [Indexed: 07/21/2024] Open
Abstract
AIMS Circular RNAs (circRNAs) are considered important regulators of biological processes, but their impact on atherosclerosis development, a key factor in coronary artery disease (CAD), has not been fully elucidated. We aimed to investigate their potential use in patients with CAD and the pathogenesis of atherosclerosis. METHODS AND RESULTS Patients with stable angina (SA) or acute coronary syndrome (ACS) and controls were selected for transcriptomic screening and quantification of circRNAs in blood cells. We stained carotid plaque samples for circRNAs and performed gain- and loss-of-function studies in vitro. Western blots, protein interaction analysis, and molecular approaches were used to perform the mechanistic study. ApoE-/- mouse models were employed in functional studies with adeno-associated virus-mediated genetic intervention. We demonstrated elevated circARCN1 expression in peripheral blood mononuclear cells from patients with SA or ACS, especially in those with ACS. Furthermore, higher circARCN1 levels were associated with a higher risk of developing SA and ACS. We also observed elevated expression of circARCN1 in carotid artery plaques. Further analysis indicated that circARCN1 was mainly expressed in monocytes and macrophages, which was also confirmed in atherosclerotic plaques. Our in vitro studies provided evidence that circARCN1 affected the interaction between HuR and ubiquitin-specific peptidase 31 (USP31) mRNA, resulting in attenuated USP31-mediated NF-κB activation. Interestingly, macrophage accumulation and inflammation in atherosclerotic plaques were markedly decreased when circARCN1 was knocked down with adeno-associated virus in macrophages of ApoE-/- mice, while circARCN1 overexpression in the model exacerbated atherosclerotic lesions. CONCLUSIONS Our findings provide solid evidence macrophagic-expressed circARCN1 plays a role in atherosclerosis development by regulating HuR-mediated USP31 mRNA stability and NF-κB activation, suggesting that circARCN1 may serve as a factor for atherosclerotic lesion formation.
Collapse
MESH Headings
- Aged
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Acute Coronary Syndrome/genetics
- Acute Coronary Syndrome/pathology
- Acute Coronary Syndrome/metabolism
- Angina, Stable/genetics
- Angina, Stable/metabolism
- Angina, Stable/pathology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Carotid Artery Diseases/genetics
- Carotid Artery Diseases/metabolism
- Carotid Artery Diseases/pathology
- Case-Control Studies
- Coronary Artery Disease/genetics
- Coronary Artery Disease/pathology
- Coronary Artery Disease/metabolism
- Disease Models, Animal
- ELAV-Like Protein 1/metabolism
- ELAV-Like Protein 1/genetics
- Gene Expression Regulation
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- NF-kappa B/metabolism
- Plaque, Atherosclerotic
- RAW 264.7 Cells
- RNA, Circular/genetics
- RNA, Circular/metabolism
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- Signal Transduction
- THP-1 Cells
- Ubiquitin Thiolesterase/genetics
- Ubiquitin Thiolesterase/metabolism
- Ubiquitin-Specific Proteases/genetics
- Ubiquitin-Specific Proteases/metabolism
Collapse
Affiliation(s)
- Zhicheng Pan
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jialan Lv
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liding Zhao
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kaidi Xing
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Runze Ye
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuesheng Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Siyuan Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hailong Yu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yangkai Lin
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruobing Li
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongfei Wang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Fang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yang Dong
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianpeng Sheng
- Zhejiang Province Key Laboratory of Pancreatic Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaolin Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, The RNA Institute, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China (UTSC), Hefei, China
| | - Ge Shan
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, The RNA Institute, School of Basic Medical Sciences, Division of Life Science and Medicine, University of Science and Technology of China (UTSC), Hefei, China
| | - Shan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Biochemistry, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongqiang Cheng
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingbo Xu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
4
|
Chen P, Li X. NLRP3 inflammasome in atherosclerosis: Mechanisms and targeted therapies. Front Pharmacol 2024; 15:1430236. [PMID: 39144618 PMCID: PMC11322363 DOI: 10.3389/fphar.2024.1430236] [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: 05/09/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Atherosclerosis (AS) is the primary pathology behind various cardiovascular diseases and the leading cause of death and disability globally. Recent evidence suggests that AS is a chronic vascular inflammatory disease caused by multiple factors. In this context, the NLRP3 inflammasome, acting as a signal transducer of the immune system, plays a critical role in the onset and progression of AS. The NLRP3 inflammasome is involved in endothelial injury, foam cell formation, and pyroptosis in AS. Therefore, targeting the NLRP3 inflammasome offers a new treatment strategy for AS. This review highlights the latest insights into AS pathogenesis and the pharmacological therapies targeting the NLRP3 inflammasome, focusing on optimal targets for small molecule inhibitors. These insights are valuable for rational drug design and the pharmacological assessment of new targeted NLRP3 inflammasome inhibitors in treating AS.
Collapse
Affiliation(s)
- Pengfei Chen
- Marine College, Shandong University, Weihai, China
| | - Xia Li
- Marine College, Shandong University, Weihai, China
- Shandong Kelun Pharmaceutical Co, Ltd., Binzhou, China
| |
Collapse
|
5
|
Völkers M, Preiss T, Hentze MW. RNA-binding proteins in cardiovascular biology and disease: the beat goes on. Nat Rev Cardiol 2024; 21:361-378. [PMID: 38163813 DOI: 10.1038/s41569-023-00958-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/06/2023] [Indexed: 01/03/2024]
Abstract
Cardiac development and function are becoming increasingly well understood from different angles, including signalling, transcriptional and epigenetic mechanisms. By contrast, the importance of the post-transcriptional landscape of cardiac biology largely remains to be uncovered, building on the foundation of a few existing paradigms. The discovery during the past decade of hundreds of additional RNA-binding proteins in mammalian cells and organs, including the heart, is expected to accelerate progress and has raised intriguing possibilities for better understanding the intricacies of cardiac development, metabolism and adaptive alterations. In this Review, we discuss the progress and new concepts on RNA-binding proteins and RNA biology and appraise them in the context of common cardiovascular clinical conditions, from cell and organ-wide perspectives. We also discuss how a better understanding of cardiac RNA-binding proteins can fill crucial knowledge gaps in cardiology and might pave the way to developing better treatments to reduce cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Mirko Völkers
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg and Mannheim, Germany
| | - Thomas Preiss
- Shine-Dalgarno Centre for RNA Innovation, John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| | - Matthias W Hentze
- European Molecular Biology Laboratory, Heidelberg, Germany.
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Germany.
| |
Collapse
|
6
|
Abdelsam SS, Ghanem SK, Zahid MA, Abunada HH, Bader L, Raïq H, Khan A, Parray A, Djouhri L, Agouni A. Human antigen R: Exploring its inflammatory response impact and significance in cardiometabolic disorders. J Cell Physiol 2024; 239:e31229. [PMID: 38426269 DOI: 10.1002/jcp.31229] [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: 11/13/2023] [Revised: 01/30/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
RNA-binding proteins (RBPs) play a crucial role in the regulation of posttranscriptional RNA networks, which can undergo dysregulation in many pathological conditions. Human antigen R (HuR) is a highly researched RBP that plays a crucial role as a posttranscriptional regulator. HuR plays a crucial role in the amplification of inflammatory signals by stabilizing the messenger RNA of diverse inflammatory mediators and key molecular players. The noteworthy correlations between HuR and its target molecules, coupled with the remarkable impacts reported on the pathogenesis and advancement of multiple diseases, position HuR as a promising candidate for therapeutic intervention in diverse inflammatory conditions. This review article examines the significance of HuR as a member of the RBP family, its regulatory mechanisms, and its implications in the pathophysiology of inflammation and cardiometabolic illnesses. Our objective is to illuminate potential directions for future research and drug development by conducting a comprehensive analysis of the existing body of research on HuR.
Collapse
Affiliation(s)
- Shahenda Salah Abdelsam
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Sarah Khalaf Ghanem
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Muhammad Ammar Zahid
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Hanan H Abunada
- Office of Vice President for Research and Graduate Studies, Qatar University, Doha, Qatar
| | - Loulia Bader
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Hicham Raïq
- Department of Social Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar
| | - Abbas Khan
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
| | - Aijaz Parray
- The Neuroscience Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Laiche Djouhri
- Department of Basic Medical Science, College of Medicine, QU health, Qatar University, Doha, Qatar
| | - Abdelali Agouni
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health, Qatar University, Doha, Qatar
- Office of Vice President for Medical & Health Sciences, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
7
|
Wardman R, Keles M, Pachkiv I, Hemanna S, Grein S, Schwarz J, Stein F, Ola R, Dobreva G, Hentze MW, Heineke J. RNA-Binding Proteins Regulate Post-Transcriptional Responses to TGF-β to Coordinate Function and Mesenchymal Activation of Murine Endothelial Cells. Arterioscler Thromb Vasc Biol 2023; 43:1967-1989. [PMID: 37650327 PMCID: PMC10521797 DOI: 10.1161/atvbaha.123.319925] [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: 07/26/2023] [Accepted: 08/18/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Endothelial cells (ECs) are primed to respond to various signaling cues. For example, TGF (transforming growth factor)-β has major effects on EC function and phenotype by driving ECs towards a more mesenchymal state (ie, triggering endothelial to mesenchymal activation), a dynamic process associated with cardiovascular diseases. Although transcriptional regulation triggered by TGF-β in ECs is well characterized, post-transcriptional regulatory mechanisms induced by TGF-β remain largely unknown. METHODS Using RNA interactome capture, we identified global TGF-β driven changes in RNA-binding proteins in ECs. We investigated specific changes in the RNA-binding patterns of hnRNP H1 (heterogeneous nuclear ribonucleoprotein H1) and Csde1 (cold shock domain containing E1) using RNA immunoprecipitation and overlapped this with RNA-sequencing data after knockdown of either protein for functional insight. Using a modified proximity ligation assay, we visualized the specific interactions between hnRNP H1 and Csde1 and target RNAs in situ both in vitro and in mouse heart sections. RESULTS Characterization of TGF-β-regulated RBPs (RNA-binding proteins) revealed hnRNP H1 and Csde1 as key regulators of the cellular response to TGF-β at the post-transcriptional level, with loss of either protein-promoting mesenchymal activation in ECs. We found that TGF-β drives an increase in binding of hnRNP H1 to its target RNAs, offsetting mesenchymal activation, but a decrease in Csde1 RNA-binding, facilitating this process. Both, hnRNP H1 and Csde1, dynamically bind and regulate specific subsets of mRNAs related to mesenchymal activation and endothelial function. CONCLUSIONS Together, we show that RBPs play a key role in the endothelial response to TGF-β stimulation at the post-transcriptional level and that the RBPs hnRNP H1 and Csde1 serve to maintain EC function and counteract mesenchymal activation. We propose that TGF-β profoundly modifies RNA-protein interaction entailing feedback and feed-forward control at the post-transcriptional level, to fine-tune mesenchymal activation in ECs.
Collapse
Affiliation(s)
- Rhys Wardman
- Department of Cardiovascular Physiology (R.W., M.K., I.P., S.H., S.G., J.H.), European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (R.W., M.K., S.H., S.G., G.D., J.H.)
| | - Merve Keles
- Department of Cardiovascular Physiology (R.W., M.K., I.P., S.H., S.G., J.H.), European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (R.W., M.K., S.H., S.G., G.D., J.H.)
| | - Ihor Pachkiv
- Department of Cardiovascular Physiology (R.W., M.K., I.P., S.H., S.G., J.H.), European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, Germany
| | - Shruthi Hemanna
- Department of Cardiovascular Physiology (R.W., M.K., I.P., S.H., S.G., J.H.), European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (R.W., M.K., S.H., S.G., G.D., J.H.)
| | - Steve Grein
- Department of Cardiovascular Physiology (R.W., M.K., I.P., S.H., S.G., J.H.), European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (R.W., M.K., S.H., S.G., G.D., J.H.)
| | - Jennifer Schwarz
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany (J.S., F.S.)
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany (J.S., F.S.)
| | - Roxana Ola
- Cardiovascular Pharmacology (R.O.), European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, Germany
| | - Gergana Dobreva
- Cardiovascular Genomics and Epigenomics (G.D.), European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (R.W., M.K., S.H., S.G., G.D., J.H.)
| | - Matthias W. Hentze
- European Molecular Biology Laboratory (EMBL), Heidelberg, Germany (M.W.H.)
| | - Joerg Heineke
- Department of Cardiovascular Physiology (R.W., M.K., I.P., S.H., S.G., J.H.), European Center for Angioscience (ECAS), Medical Faculty Mannheim of Heidelberg University, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim (R.W., M.K., S.H., S.G., G.D., J.H.)
| |
Collapse
|
8
|
Sachse M, Tual-Chalot S, Ciliberti G, Amponsah-Offeh M, Stamatelopoulos K, Gatsiou A, Stellos K. RNA-binding proteins in vascular inflammation and atherosclerosis. Atherosclerosis 2023; 374:55-73. [PMID: 36759270 DOI: 10.1016/j.atherosclerosis.2023.01.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/01/2022] [Accepted: 01/12/2023] [Indexed: 01/19/2023]
Abstract
Atherosclerotic cardiovascular disease (ASCVD) remains the major cause of premature death and disability worldwide, even when patients with an established manifestation of atherosclerotic heart disease are optimally treated according to the clinical guidelines. Apart from the epigenetic control of transcription of the genetic information to messenger RNAs (mRNAs), gene expression is tightly controlled at the post-transcriptional level before the initiation of translation. Although mRNAs are traditionally perceived as the messenger molecules that bring genetic information from the nuclear DNA to the cytoplasmic ribosomes for protein synthesis, emerging evidence suggests that processes controlling RNA metabolism, driven by RNA-binding proteins (RBPs), affect cellular function in health and disease. Over the recent years, vascular endothelial cell, smooth muscle cell and immune cell RBPs have emerged as key co- or post-transcriptional regulators of several genes related to vascular inflammation and atherosclerosis. In this review, we provide an overview of cell-specific function of RNA-binding proteins involved in all stages of ASCVD and how this knowledge may be used for the development of novel precision medicine therapeutics.
Collapse
Affiliation(s)
- Marco Sachse
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Department of Cardiovascular Surgery, University Heart Center, University Hospital Hamburg Eppendorf, Hamburg, Germany
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.
| | - Giorgia Ciliberti
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany
| | - Michael Amponsah-Offeh
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Aikaterini Gatsiou
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Konstantinos Stellos
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site, Mannheim, Germany; Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany.
| |
Collapse
|
9
|
Bibli SI, Fleming I. Oxidative Post-Translational Modifications: A Focus on Cysteine S-Sulfhydration and the Regulation of Endothelial Fitness. Antioxid Redox Signal 2021; 35:1494-1514. [PMID: 34346251 DOI: 10.1089/ars.2021.0162] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Changes in the oxidative balance can affect cellular physiology and adaptation through redox signaling. The endothelial cells that line blood vessels are particularly sensitive to reactive oxygen species, which can alter cell function by a number of mechanisms, including the oxidative post-translational modification (oxPTM) of proteins on critical cysteine thiols. Such modifications can act as redox-switches to alter the function of targeted proteins. Recent Advances: Mapping the cysteine oxPTM proteome and characterizing the effects of individual oxPTMs to gain insight into consequences for cellular responses has proven challenging. A recent addition to the list of reversible oxPTMs that contributes to cellular redox homeostasis is persulfidation or S-sulfhydration. Critical Issues: It has been estimated that up to 25% of proteins are S-sulfhydrated, making this modification almost as abundant as phosphorylation. In the endothelium, persulfides are generated by the trans-sulfuration pathway that catabolizes cysteine and cystathionine to generate hydrogen sulfide (H2S) and H2S-related sulfane sulfur compounds (H2Sn). This pathway is of particular importance for the vascular system, as the enzyme cystathionine γ lyase (CSE) in endothelial cells accounts for a significant portion of total vascular H2S/H2Sn production. Future Directions: Impaired CSE activity in endothelial dysfunction has been linked with marked changes in the endothelial cell S-sulfhydrome and can contribute to the development of atherosclerosis and hypertension. It will be interesting to determine how changes in the S-sulfhydration of specific networks of proteins contribute to endothelial cell physiology and pathophysiology. Antioxid. Redox Signal. 35, 1494-1514.
Collapse
Affiliation(s)
- Sofia-Iris Bibli
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt am Main, Germany
| |
Collapse
|
10
|
Ding Y, Yin R, Zhang S, Xiao Q, Zhao H, Pan X, Zhu X. The Combined Regulation of Long Non-coding RNA and RNA-Binding Proteins in Atherosclerosis. Front Cardiovasc Med 2021; 8:731958. [PMID: 34796209 PMCID: PMC8592911 DOI: 10.3389/fcvm.2021.731958] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/07/2021] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is a complex disease closely related to the function of endothelial cells (ECs), monocytes/macrophages, and vascular smooth muscle cells (VSMCs). Despite a good understanding of the pathogenesis of atherosclerosis, the underlying molecular mechanisms are still only poorly understood. Therefore, atherosclerosis continues to be an important clinical issue worthy of further research. Recent evidence has shown that long non-coding RNAs (lncRNAs) and RNA-binding proteins (RBPs) can serve as important regulators of cellular function in atherosclerosis. Besides, several studies have shown that lncRNAs are partly dependent on the specific interaction with RBPs to exert their function. This review summarizes the important contributions of lncRNAs and RBPs in atherosclerosis and provides novel and comprehensible interaction models of lncRNAs and RBPs.
Collapse
Affiliation(s)
- Yuanyuan Ding
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruihua Yin
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shuai Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qi Xiao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongqin Zhao
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoyan Zhu
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
11
|
Si R, Cabrera JTO, Tsuji-Hosokawa A, Guo R, Watanabe M, Gao L, Lee YS, Moon JS, Scott BT, Wang J, Ashton AW, Rao JN, Wang JY, Yuan JXJ, Makino A. HuR/Cx40 downregulation causes coronary microvascular dysfunction in type 2 diabetes. JCI Insight 2021; 6:147982. [PMID: 34747371 PMCID: PMC8663561 DOI: 10.1172/jci.insight.147982] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 09/29/2021] [Indexed: 11/30/2022] Open
Abstract
Patients with diabetes with coronary microvascular disease (CMD) exhibit higher cardiac mortality than patients without CMD. However, the molecular mechanism by which diabetes promotes CMD is poorly understood. RNA-binding protein human antigen R (HuR) is a key regulator of mRNA stability and translation; therefore, we investigated the role of HuR in the development of CMD in mice with type 2 diabetes. Diabetic mice exhibited decreases in coronary flow velocity reserve (CFVR; a determinant of coronary microvascular function) and capillary density in the left ventricle. HuR levels in cardiac endothelial cells (CECs) were significantly lower in diabetic mice and patients with diabetes than the controls. Endothelial-specific HuR-KO mice also displayed significant reductions in CFVR and capillary density. By examining mRNA levels of 92 genes associated with endothelial function, we found that HuR, Cx40, and Nox4 levels were decreased in CECs from diabetic and HuR-KO mice compared with control mice. Cx40 expression and HuR binding to Cx40 mRNA were downregulated in CECs from diabetic mice. Cx40-KO mice exhibited decreased CFVR and capillary density, whereas endothelium-specific Cx40 overexpression increased capillary density and improved CFVR in diabetic mice. These data suggest that decreased HuR contributes to the development of CMD in diabetes through downregulation of gap junction protein Cx40 in CECs.
Collapse
Affiliation(s)
- Rui Si
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA.,Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Shaanxi, China
| | | | | | - Rui Guo
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA
| | - Makiko Watanabe
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA
| | - Lei Gao
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Yun Sok Lee
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Jae-Su Moon
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Brian T Scott
- Department of Medicine, UCSD, La Jolla, California, USA
| | - Jian Wang
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Anthony W Ashton
- Division of Perinatal Research, Kolling Institute of Medical Research, University of Sydney, New South Wales, Australia
| | - Jaladanki N Rao
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jian-Ying Wang
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Ayako Makino
- Department of Physiology, The University of Arizona (UA), Tucson, Arizona, USA.,Department of Medicine, UCSD, La Jolla, California, USA
| |
Collapse
|
12
|
Chen W, Li L, Wang J, Li Q, Zhang R, Wang S, Wu Y, Xing D. Extracellular vesicle YRNA in atherosclerosis. Clin Chim Acta 2021; 517:15-22. [DOI: 10.1016/j.cca.2021.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/04/2021] [Accepted: 02/05/2021] [Indexed: 02/08/2023]
|
13
|
Hepatic HuR modulates lipid homeostasis in response to high-fat diet. Nat Commun 2020; 11:3067. [PMID: 32546794 PMCID: PMC7298042 DOI: 10.1038/s41467-020-16918-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 05/26/2020] [Indexed: 12/23/2022] Open
Abstract
Lipid transport and ATP synthesis are critical for the progression of non-alcoholic fatty liver disease (NAFLD), but the underlying mechanisms are largely unknown. Here, we report that the RNA-binding protein HuR (ELAVL1) forms complexes with NAFLD-relevant transcripts. It associates with intron 24 of Apob pre-mRNA, with the 3′UTR of Uqcrb, and with the 5′UTR of Ndufb6 mRNA, thereby regulating the splicing of Apob mRNA and the translation of UQCRB and NDUFB6. Hepatocyte-specific HuR knockout reduces the expression of APOB, UQCRB, and NDUFB6 in mice, reducing liver lipid transport and ATP synthesis, and aggravating high-fat diet (HFD)-induced NAFLD. Adenovirus-mediated re-expression of HuR in hepatocytes rescues the effect of HuR knockout in HFD-induced NAFLD. Our findings highlight a critical role of HuR in regulating lipid transport and ATP synthesis. Human antigen R (HuR) is a RNA binding protein involved in the regulation of many cellular functions. Here the authors show that, hepatocyte specific deletion of HuR exacerbates high-fat diet-induced NAFLD in mice by regulating transcripts involved in lipid transport and ATP synthesis.
Collapse
|
14
|
Schultz CW, Preet R, Dhir T, Dixon DA, Brody JR. Understanding and targeting the disease-related RNA binding protein human antigen R (HuR). WILEY INTERDISCIPLINARY REVIEWS-RNA 2020; 11:e1581. [PMID: 31970930 DOI: 10.1002/wrna.1581] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/02/2019] [Accepted: 12/07/2019] [Indexed: 02/06/2023]
Abstract
Altered gene expression is a characteristic feature of many disease states such as tumorigenesis, and in most cancers, it facilitates cancer cell survival and adaptation. Alterations in global gene expression are strongly impacted by post-transcriptional gene regulation. The RNA binding protein (RBP) HuR (ELAVL1) is an established regulator of post-transcriptional gene regulation and is overexpressed in most human cancers. In many cancerous settings, HuR is not only overexpressed, but it is "overactive" as denoted by increased subcellular localization within the cytoplasm. This dysregulation of HuR expression and cytoplasmic localization allows HuR to stabilize and increase the translation of various prosurvival messenger RNA (mRNAs) involved in the pathogenesis of numerous cancers and various diseases. Based on almost 20 years of work, HuR is now recognized as a therapeutic target. Herein, we will review the role HuR plays in the pathophysiology of different diseases and ongoing therapeutic strategies to target HuR. We will focus on three ongoing-targeted strategies: (1) inhibiting HuR's translocation from the nucleus to the cytoplasm; (2) inhibiting the ability of HuR to bind target RNA; and (3) silencing HuR expression levels. In an oncologic setting, HuR has been demonstrated to be critical for a cancer cell's ability to survive a variety of cancer relevant stressors (including drugs and elements of the tumor microenvironment) and targeting this protein has been shown to sensitize cancer cells further to insult. We strongly believe that targeting HuR could be a powerful therapeutic target to treat different diseases, particularly cancer, in the near future. This article is categorized under: RNA in Disease and Development > RNA in Disease NRA Turnover and Surveillance > Regulation of RNA Stability Translation > Translation Regulation.
Collapse
Affiliation(s)
- Christopher W Schultz
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Ranjan Preet
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas
| | - Teena Dhir
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Dan A Dixon
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas
| | - Jonathan R Brody
- Department of Surgery, Jefferson Pancreas, Biliary and Related Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Li W, Zhi W, Zhao J, Li W, Zang L, Liu F, Niu X. Cinnamaldehyde attenuates atherosclerosis via targeting the IκB/NF-κB signaling pathway in high fat diet-induced ApoE−/− mice. Food Funct 2019; 10:4001-4009. [DOI: 10.1039/c9fo00396g] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cinnamaldehyde is a flavor isolated from the bark of Cinnamomum cassia Presl and exerts anti-inflammation effects in various diseases.
Collapse
Affiliation(s)
- Weifeng Li
- School of Pharmacy
- Xi'an Jiaotong University
- Xi'an 710061
- P.R. China
| | - Wenbing Zhi
- School of Pharmacy
- Xi'an Jiaotong University
- Xi'an 710061
- P.R. China
- Shaanxi Academy of Traditional Chinese Medicine
| | - Jinmeng Zhao
- School of Pharmacy
- Xi'an Jiaotong University
- Xi'an 710061
- P.R. China
| | - Wenqi Li
- School of Pharmacy
- Xi'an Jiaotong University
- Xi'an 710061
- P.R. China
| | - Lulu Zang
- School of Pharmacy
- Xi'an Jiaotong University
- Xi'an 710061
- P.R. China
| | - Fang Liu
- School of Pharmacy
- Xi'an Jiaotong University
- Xi'an 710061
- P.R. China
| | - Xiaofeng Niu
- School of Pharmacy
- Xi'an Jiaotong University
- Xi'an 710061
- P.R. China
| |
Collapse
|