1
|
Qiu Y, Xu Q, Xie P, He C, Li Q, Yao X, Mao Y, Wu X, Zhang T. Epigenetic modifications and emerging therapeutic targets in cardiovascular aging and diseases. Pharmacol Res 2025; 211:107546. [PMID: 39674563 DOI: 10.1016/j.phrs.2024.107546] [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: 11/12/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
The complex mechanisms underlying the development of cardiovascular diseases remain not fully elucidated. Epigenetics, which modulates gene expression without DNA sequence changes, is shedding light on these mechanisms and their heritable effects. This review focus on epigenetic regulation in cardiovascular aging and diseases, detailing specific epigenetic enzymes such as DNA methyltransferases (DNMTs), histone acetyltransferases (HATs), and histone deacetylases (HDACs), which serve as writers or erasers that modify the epigenetic landscape. We also discuss the readers of these modifications, such as the 5-methylcytosine binding domain proteins, and the erasers ten-eleven translocation (TET) proteins. The emerging role of RNA methylation, particularly N6-methyladenosine (m6A), in cardiovascular pathogenesis is also discussed. We summarize potential therapeutic targets, such as key enzymes and their inhibitors, including DNMT inhibitors like 5-azacytidine and decitabine, HDAC inhibitors like belinostat and givinotide, some of which have been approved by the FDA for various malignancies, suggesting their potential in treating cardiovascular diseases. Furthermore, we highlight the role of novel histone modifications and their associated enzymes, which are emerging as potential therapeutic targets in cardiovascular diseases. Thus, by incorporating the recent studies involving patients with cardiovascular aging and diseases, we aim to provide a more detailed and updated review that reflects the advancements in the field of epigenetic modification in cardiovascular diseases.
Collapse
Affiliation(s)
- Yurou Qiu
- GMU-GIBH Joint School of Life Sciences, Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, The Sixth School of Clinical Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Qing Xu
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Peichen Xie
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Chenshuang He
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Qiuchan Li
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Xin Yao
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Yang Mao
- 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, Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Xiaoqian Wu
- 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, Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| | - Tiejun Zhang
- GMU-GIBH Joint School of Life Sciences, Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, the Second Affiliated Hospital, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, State Key Laboratory of Respiratory Disease, The Sixth School of Clinical Medicine, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
2
|
Pan X, Song Y, Liang Y, Feng G, Wang Z. Roseburia intestinalis: A possible target for vascular calcification. Heliyon 2024; 10:e39865. [PMID: 39524709 PMCID: PMC11550659 DOI: 10.1016/j.heliyon.2024.e39865] [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: 01/21/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
With the advancement of metagenomics and metabolomics techniques, the crucial role of the gut microbiome in intestinal, cardiovascular, and metabolic disorders has been extensively explored. Vascular calcification (VC) is common in atherosclerosis, hypertension, diabetes mellitus, and chronic kidney disease. Moreover, it is a significant cause of cardiovascular diseases and mortality. Roseburia intestinalis, as a promising candidate for the next generation of probiotics, plays a substantial role in inhibiting the systemic inflammatory response and holds great potential in the treatment of intestinal diseases, cardiovascular diseases, and metabolic disorders. Its primary metabolite, butyrate, acts on specific receptors (GPR43, GPR41, GPR109a). It enters cells via transporters (MCT1, SMCT1), affecting gene expression through HDACs, PPARγ and Nrf2, promoting energy metabolism and changing the concentration of other metabolites (including AGEs, LPS, BHB) in the circulation to affect the body's life activities. In this paper, we focus on the possible mechanism of the primary metabolite butyrate of Roseburia intestinalis in inhibiting VC, which may become a potential therapeutic target for the treatment of VC and the ways to enhance its effect.
Collapse
Affiliation(s)
- Xinyun Pan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yunjian Song
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yapeng Liang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guoquan Feng
- Department of Imaging, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| |
Collapse
|
3
|
Zhu XX, Meng XY, Chen G, Su JB, Fu X, Xu AJ, Liu Y, Hou XH, Qiu HB, Sun QY, Hu JY, Lv ZL, Sun HJ, Jiang HB, Han ZJ, Zhu J, Lu QB. Nesfatin-1 enhances vascular smooth muscle calcification through facilitating BMP-2 osteogenic signaling. Cell Commun Signal 2024; 22:488. [PMID: 39394127 PMCID: PMC11468037 DOI: 10.1186/s12964-024-01873-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024] Open
Abstract
Vascular calcification (VC) arises from the accumulation of calcium salts in the intimal or tunica media layer of the aorta, contributing to higher risk of cardiovascular events and mortality. Despite this, the mechanisms driving VC remain incompletely understood. We previously described that nesfatin-1 functioned as a switch for vascular smooth muscle cells (VSMCs) plasticity in hypertension and neointimal hyperplasia. In this study, we sought to investigate the role and mechanism of nesfatin-1 in VC. The expression of nesfatin-1 was measured in calcified VSMCs and aortas, as well as in patients. Loss- and gain-of-function experiments were evaluated the roles of nesfatin-1 in VC pathogenesis. The transcription activation of nesfatin-1 was detected using a mass spectrometry. We found higher levels of nesfatin-1 in both calcified VSMCs and aortas, as well as in patients with coronary calcification. Loss-of-function and gain-of-function experiments revealed that nesfatin-1 was a key regulator of VC by facilitating the osteogenic transformation of VSMCs. Mechanistically, nesfatin-1 promoted the de-ubiquitination and stability of BMP-2 via inhibiting the E3 ligase SYTL4, and the interaction of nesfatin-1 with BMP-2 potentiated BMP-2 signaling and induced phosphorylation of Smad, followed by HDAC4 phosphorylation and nuclear exclusion. The dissociation of HDAC4 from RUNX2 elicited RUNX2 acetylation and subsequent nuclear translocation, leading to the transcription upregulation of OPN, a critical player in VC. From a small library of natural compounds, we identified that Curculigoside and Chebulagic acid reduced VC development via binding to and inhibiting nesfatin-1. Eventually, we designed a mass spectrometry-based DNA-protein interaction screening to identify that STAT3 mediated the transcription activation of nesfatin-1 in the context of VC. Overall, our study demonstrates that nesfatin-1 enhances BMP-2 signaling by inhibiting the E3 ligase SYTL4, thereby stabilizing BMP-2 and facilitating the downstream phosphorylation of SMAD1/5/9 and HDAC4. This signaling cascade leads to RUNX2 activation and the transcriptional upregulation of MSX2, driving VC. These insights position nesfatin-1 as a potential therapeutic target for preventing or treating VC, advancing our understanding of the molecular mechanisms underlying this critical cardiovascular condition.
Collapse
Affiliation(s)
- Xue-Xue Zhu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
- Department of Physiology, Eberhard-Karls-University of Tübingen, Tübingen University, Tübingen, 72076, Germany
| | - Xin-Yu Meng
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Guo Chen
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Jia-Bao Su
- Department of Anesthesiology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214122, China
| | - Xiao Fu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - An-Jing Xu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yao Liu
- Department of Ultrasound, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Xiao-Hui Hou
- Department of Ultrasound, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, China
| | - Hong-Bo Qiu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Qing-Yi Sun
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Jin-Yi Hu
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Zhuo-Lin Lv
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Hai-Jian Sun
- MOE Medical Basic Research Innovation Center for Gut Microbiota and Chronic Diseases, School of Medicine, Jiangnan University, Wuxi, 214122, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Hai-Bin Jiang
- Department of Cardiology, Wuxi No.2 People's Hospital (Jiangnan University Medical Center), Wuxi School of Medicine, Jiangnan University, Wuxi, 214001, China.
| | - Zhi-Jun Han
- Department of Clinical Research Center, Jiangnan University Medical Center (Wuxi No.2 People's Hospital), Wuxi School of Medicine, Jiangnan University, Wuxi, 214001, China.
| | - Jian Zhu
- Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214122, China.
| | - Qing-Bo Lu
- Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
4
|
Liang X, Li Y, Wang P, Liu H. Key regulators of vascular calcification in chronic kidney disease: Hyperphosphatemia, BMP2, and RUNX2. PeerJ 2024; 12:e18063. [PMID: 39308809 PMCID: PMC11416758 DOI: 10.7717/peerj.18063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
Vascular calcification is quite common in patients with end-stage chronic kidney disease and is a major trigger for cardiovascular complications in these patients. These complications significantly impact the survival rate and long-term prognosis of individuals with chronic kidney disease. Numerous studies have demonstrated that the development of vascular calcification involves various pathophysiological mechanisms, with the osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) being of utmost importance. High phosphate levels, bone morphogenetic protein 2 (BMP2), and runt-related transcription factor 2 (RUNX2) play crucial roles in the osteogenic transdifferentiation process of VSMCs. This article primarily reviews the molecular mechanisms by which high phosphate, BMP2, and RUNX2 regulate vascular calcification secondary to chronic kidney disease, and discusses the complex interactions among these factors and their impact on the progression of vascular calcification. The insights provided here aim to offer new perspectives for future research on the phenotypic switching and osteogenic transdifferentiation of VSMCs, as well as to aid in optimizing clinical treatment strategies for this condition, bearing significant clinical and scientific implications.
Collapse
Affiliation(s)
- Xinhua Liang
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Zhanjiang, Guangdong Province, China
| | - Yankun Li
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Zhanjiang, Guangdong Province, China
| | - Peng Wang
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, Guangdong, China
| | - Huafeng Liu
- Affiliated Hospital of Guangdong Medical University, Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Zhanjiang, Guangdong, China
| |
Collapse
|
5
|
Shi F. Understanding the roles of salt-inducible kinases in cardiometabolic disease. Front Physiol 2024; 15:1426244. [PMID: 39081779 PMCID: PMC11286596 DOI: 10.3389/fphys.2024.1426244] [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: 05/01/2024] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
Salt-inducible kinases (SIKs) are serine/threonine kinases of the adenosine monophosphate-activated protein kinase family. Acting as mediators of a broad array of neuronal and hormonal signaling pathways, SIKs play diverse roles in many physiological and pathological processes. Phosphorylation by the upstream kinase liver kinase B1 is required for SIK activation, while phosphorylation by protein kinase A induces the binding of 14-3-3 protein and leads to SIK inhibition. SIKs are subjected to auto-phosphorylation regulation and their activity can also be modulated by Ca2+/calmodulin-dependent protein kinase in response to cellular calcium influx. SIKs regulate the physiological processes through direct phosphorylation on various substrates, which include class IIa histone deacetylases, cAMP-regulated transcriptional coactivators, phosphatase methylesterase-1, among others. Accumulative body of studies have demonstrated that SIKs are important regulators of the cardiovascular system, including early works establishing their roles in sodium sensing and vascular homeostasis and recent progress in pulmonary arterial hypertension and pathological cardiac remodeling. SIKs also regulate inflammation, fibrosis, and metabolic homeostasis, which are essential pathological underpinnings of cardiovascular disease. The development of small molecule SIK inhibitors provides the translational opportunity to explore their potential as therapeutic targets for treating cardiometabolic disease in the future.
Collapse
Affiliation(s)
- Fubiao Shi
- Department of Medicine, Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
6
|
Liu R, Li L, Wang Z, Zhu J, Ji Y. Acetylated Histone Modifications: Intersection of Diabetes and Atherosclerosis. J Cardiovasc Pharmacol 2024; 83:207-219. [PMID: 37989137 DOI: 10.1097/fjc.0000000000001516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
ABSTRACT Worldwide, type 2 diabetes is predominant form of diabetes, and it is mainly affected by the environment. Furthermore, the offspring of patients with type 2 diabetes and metabolic disorder syndrome may have a higher risk of diabetes and cardiovascular disease, which indicates that the environmental impact on diabetes prevalence can be transmitted across generations. In the process of diabetes onset and intergenerational transmission, the genetic structure of the individual is not directly changed but is regulated by epigenetics. In this process, genes or histones are modified, resulting in selective expression of proteins. This modification will affect not only the onset of diabetes but also the related onset of atherosclerosis. Acetylation and deacetylation may be important regulatory factors for the above lesions. Therefore, in this review, based on the whole process of atherosclerosis evolution, we explored the possible existence of acetylation/deacetylation caused by diabetes. However, because of the lack of atherosclerosis-related acetylation studies directly based on diabetic models, we also used a small number of experiments involving nondiabetic models of related molecular mechanisms.
Collapse
Affiliation(s)
| | | | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China; and
| | - Jie Zhu
- Department of Cardiology, The Lu'an Hospital Affiliated to Anhui Medical University, The Lu' an People's Hospital, Lu'an, China
| | | |
Collapse
|
7
|
Helms RS, Marin-Gonzalez A, Patel CH, Sun IH, Wen J, Leone RD, Duvall B, Gao RD, Ha T, Tsukamoto T, Slusher BS, Pomerantz JL, Powell JD. SIKs Regulate HDAC7 Stabilization and Cytokine Recall in Late-Stage T Cell Effector Differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1767-1782. [PMID: 37947442 PMCID: PMC10842463 DOI: 10.4049/jimmunol.2300248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 10/05/2023] [Indexed: 11/12/2023]
Abstract
Understanding the mechanisms underlying the acquisition and maintenance of effector function during T cell differentiation is important to unraveling how these processes can be dysregulated in the context of disease and manipulated for therapeutic intervention. In this study, we report the identification of a previously unappreciated regulator of murine T cell differentiation through the evaluation of a previously unreported activity of the kinase inhibitor, BioE-1197. Specifically, we demonstrate that liver kinase B1 (LKB1)-mediated activation of salt-inducible kinases epigenetically regulates cytokine recall potential in effector CD8+ and Th1 cells. Evaluation of this phenotype revealed that salt-inducible kinase-mediated phosphorylation-dependent stabilization of histone deacetylase 7 (HDAC7) occurred during late-stage effector differentiation. HDAC7 stabilization increased nuclear HDAC7 levels, which correlated with total and cytokine loci-specific reductions in the activating transcription mark histone 3 lysine 27 acetylation (H3K27Ac). Accordingly, HDAC7 stabilization diminished transcriptional induction of cytokine genes upon restimulation. Inhibition of this pathway during differentiation produced effector T cells epigenetically poised for enhanced cytokine recall. This work identifies a previously unrecognized target for enhancing effector T cell functionality.
Collapse
Affiliation(s)
- Rachel S Helms
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alberto Marin-Gonzalez
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Boston, MA
| | - Chirag H Patel
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
- Calico Life Sciences LLC, South San Francisco, CA
| | - Im-Hong Sun
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Surgery, University of California San Francisco, San Francisco, CA
| | - Jiayu Wen
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Robert D Leone
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Run-Duo Gao
- Johns Hopkins Drug Discovery, Baltimore, MD
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Taekjip Ha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Boston, MA
| | - Takashi Tsukamoto
- Johns Hopkins Drug Discovery, Baltimore, MD
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Baltimore, MD
- Department of Neurology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joel L Pomerantz
- Department of Biological Chemistry, Institute for Cell Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jonathan D Powell
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney-Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
- Calico Life Sciences LLC, South San Francisco, CA
| |
Collapse
|
8
|
Zhang L, Xia C, Yang Y, Sun F, Zhang Y, Wang H, Liu R, Yuan M. DNA methylation and histone post-translational modifications in atherosclerosis and a novel perspective for epigenetic therapy. Cell Commun Signal 2023; 21:344. [PMID: 38031118 PMCID: PMC10688481 DOI: 10.1186/s12964-023-01298-8] [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: 08/08/2022] [Accepted: 08/27/2023] [Indexed: 12/01/2023] Open
Abstract
Atherosclerosis, which is a vascular pathology characterized by inflammation and plaque build-up within arterial vessel walls, acts as the important cause of most cardiovascular diseases. Except for a lipid-depository and chronic inflammatory, increasing evidences propose that epigenetic modifications are increasingly associated with atherosclerosis and are of interest from both therapeutic and biomarker perspectives. The chronic progressive nature of atherosclerosis has highlighted atherosclerosis heterogeneity and the fact that specific cell types in the complex milieu of the plaque are, by far, not the only initiators and drivers of atherosclerosis. Instead, the ubiquitous effects of cell type are tightly controlled and directed by the epigenetic signature, which, in turn, is affected by many proatherogenic stimuli, including low-density lipoprotein, proinflammatory, and physical forces of blood circulation. In this review, we summarize the role of DNA methylation and histone post-translational modifications in atherosclerosis. The future research directions and potential therapy for the management of atherosclerosis are also discussed. Video Abstract.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Chenhai Xia
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Yongjun Yang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Fangfang Sun
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Yu Zhang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Huan Wang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Rui Liu
- Department of Rehabilitation, Tangdu Hospital, Air Force Military Medical University, No. 1 Xinsi Road, Xi'an 710000, China.
| | - Ming Yuan
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
9
|
Monisha K, Mahema S, Chokkalingam M, Ahmad SF, Emran TB, Prabu P, Ahmed SSSJ. Elucidating the Histone Deacetylase Gene Expression Signatures in Peripheral Blood Mononuclear Cells That Correlate Essential Cardiac Function and Aid in Classifying Coronary Artery Disease through a Logistic Regression Model. Biomedicines 2023; 11:2952. [PMID: 38001953 PMCID: PMC10669643 DOI: 10.3390/biomedicines11112952] [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: 09/30/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/26/2023] Open
Abstract
A proinflammatory role of HDACs has been implicated in the pathogenesis of atherosclerosis as an emerging novel epigenetic diagnostic biomarker. However, its association with the clinical and cardiovascular function in coronary artery disease is largely unknown. The study aimed to profile the gene expression of HDAC1-11 in human peripheral blood mononuclear cells and to evaluate their influence on hematological, biochemical, and two-dimensional echocardiographic indices in CAD. The HDAC gene expression profiles were assessed in 62 angioproven CAD patients and compared with 62 healthy controls. Among the HDACs, upregulated HDACs 1,2, 4, 6, 8, 9, and 11 were upregulated, and HDAC3 was downregulated, which was significantly (p ≤ 0.05) linked with the hematological (basophils, lymphocytes, monocytes, and neutrophils), biochemical (LDL, HDL, and TGL), and echocardiographic parameters (cardiac function: biplane LVEF, GLS, MV E/A, IVRT, and PV S/D) in CAD. Furthermore, our constructed diagnostic model with the crucial HDACs establishes the most crucial HDACs in the classification of CAD from control with an excellent accuracy of 88.6%. Conclusively, our study has provided a novel perspective on the HDAC gene expression underlying cardiac function that is useful in developing molecular methods for CAD diagnosis.
Collapse
Affiliation(s)
- K. Monisha
- Drug Discovery and Multi-omics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam 603103, India
| | - S. Mahema
- Drug Discovery and Multi-omics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam 603103, India
| | - M. Chokkalingam
- Department of Cardiology, Chettinad Hospital and Research Institute, Chettinad Health City, Kelambakkam 603103, India
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Talha Bin Emran
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Paramasivam Prabu
- Madras Diabetes Research Foundation, Chennai 600086, India
- Department of Neurology, University of New Mexico Albuquerque, Albuquerque, NM 87131, USA
| | - Shiek S. S. J. Ahmed
- Drug Discovery and Multi-omics Laboratory, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Kelambakkam 603103, India
| |
Collapse
|
10
|
Wang K, Geng B, Shen Q, Wang Y, Shi J, Dong N. Global, regional, and national incidence, mortality, and disability-adjusted life years of non-rheumatic valvular heart disease and trend analysis from 1990 to 2019: Results from the Global Burden of Disease study 2019. Asian Cardiovasc Thorac Ann 2023; 31:706-722. [PMID: 37674443 DOI: 10.1177/02184923231200695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
BACKGROUND In the context of the population growing and aging worldwide, the incidence of non-rheumatic valvular heart disease increased rapidly. This study aimed to describe the burden of non-rheumatic valvular heart disease, providing an up-to-date and comprehensive analysis on the global and regional levels and time trends from 1900 to 2019. METHODS The Global Burden of Disease 2019 was used to obtain data for this analysis. Non-rheumatic valvular heart disease in the Global Burden of Disease study includes both non-rheumatic calcific aortic valve disease and non-rheumatic degenerative mitral valve disease. The incidence, mortality, and disability-adjusted life year in 204 countries from 1990 to 2019 were analyzed by location, year, sex, age, and socio-demographic index. Estimated annual percentage change was calculated to represent the temporal trends from 1990 to 2019. Spearman's rank order correlation was used to determine the correlation between socio-demographic index and the incidence and burden of non-rheumatic valvular heart disease. RESULTS Globally, there were 1.65 million (95% uncertainty interval, 1.56-1.76 million) incident cases, 0.16 million (95% uncertainty interval, 0.14-0.18 million) death cases, and 2.79 million (95% uncertainty interval, 2.52-3.31 million) disability-adjusted life years of non-rheumatic valvular heart disease. Compared with 1990, the number of incident cases, death cases, and disability-adjusted life years in 2019 increased by 104.58%, 210.60%, and 167.62%, respectively, the age-standardized incidence rate (estimated annual percentage change, 0.39; 95% confidence interval, 0.29 to 0.49) increased due to population growth, and the age-standardized death rates (estimated annual percentage change, -0.32; 95% confidence interval, -0.39 to -0.25) and age-standardized disability-adjusted life year rate (estimated annual percentage change, -0.81; 95% confidence interval, -0.87 to -0.74) decreased during this period. Regarding the socio-demographic index, the highest age-standardized incidence, death, and disability-adjusted life year rates of non-rheumatic valvular heart disease were found in high-socio-demographic index countries in 2019. Meantime, the age-standardized incidence rate remained increased from 1990 to 2019, while significant decreases were found in the age-standardized death rate and age-standardized disability-adjusted life year rate. Females have higher age-standardized incidence rate, while higher age-standardized death rate and age-standardized disability-adjusted life year rate belong to males globally during the period of 1990-2019. Increasing trends were observed for both incidence, death, and disability-adjusted life year rates with age. High systolic blood pressure was the leading cause for non-rheumatic valvular heart disease across all ages. CONCLUSIONS From 1990 to 2019, the age-standardized incidence rate of non-rheumatic valvular heart disease remained increased, while age-standardized death rate and age-standardized disability-adjusted life year rate decreased, resulting from the growing population worldwide and improving medical resources. The aged, who has high systolic blood pressure and diet high in sodium, should pay more attention to, especially in high-socio-demographic index regions. With the population aging, the number of patients who require heart valve replacement is estimated to increase significantly in the future. Effective measures are warranted to control and treat the incidence and burden of non-rheumatic valvular heart disease.
Collapse
Affiliation(s)
- Kan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Bingchuan Geng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiang Shen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - JiaWei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - NianGuo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
11
|
Huang J, Fan H, Chen YM, Wang CN, Guan W, Li WY, Shi TS, Chen WJ, Zhu BL, Liu JF, Jiang B. The salt-inducible kinases inhibitor HG-9-91-01 exhibits antidepressant-like actions in mice exposed to chronic unpredictable mild stress. Neuropharmacology 2023; 227:109437. [PMID: 36702294 DOI: 10.1016/j.neuropharm.2023.109437] [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: 08/23/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/24/2023]
Abstract
Major depressive disorder is a frequently occurring neuropsychiatric disorder throughout the world. However, the limited and delayed therapeutic efficacy of monoaminergic medications has led to intensive research efforts to develop novel antidepressants. We have previously demonstrated that hippocampal salt-inducible kinase 2 (SIK2) plays a role in the pathogenesis of depression via regulating the downstream CREB-regulated transcription coactivator 1 (CRTC1)-cAMP response element-binding protein (CREB)-brain derived neurotrophic factor (BDNF) pathway. HG-9-91-01 is a potent and selective inhibitor of salt-inducible kinases (SIKs). The present study aims to explore whether HG-9-91-01 has antidepressant-like actions in male C57BL/6J mice. The chronic unpredictable mild stress (CUMS) model of depression, various behavioral tests, western blotting, co-immunoprecipitation, immunofluorescence, stereotactic infusion, and viral-mediated genetic knockdown were used together. It was found that hippocampal infusion of HG-9-91-01 induced significant antidepressant-like effects in the CUMS model, accompanied with preventing the enhancement of CUMS on the hippocampal SIK2 expression and cytoplasmic translocation of CRTC1. HG-9-91-01 treatment also reversed the decreasing effects of CUMS on the BDNF signaling cascade and adult neurogenesis in the hippocampus. Moreover, the antidepressant-like actions of HG-9-91-01 in mice required the hippocampal CRTC1-CREB-BDNF pathway. In conclusion, HG-9-91-01 has potential of being a novel antidepressant candidate.
Collapse
Affiliation(s)
- Jie Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Hua Fan
- The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yan-Mei Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Cheng-Niu Wang
- Basic Medical Research Centre, Medical College, Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei-Yu Li
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Tian-Shun Shi
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Wei-Jia Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Bao-Lun Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Jian-Feng Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China.
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
12
|
Salt-inducible kinase 1 deficiency promotes vascular remodeling in pulmonary arterial hypertension via enhancement of yes-associated protein-mediated proliferation. Heliyon 2022; 8:e11016. [PMID: 36276742 PMCID: PMC9582722 DOI: 10.1016/j.heliyon.2022.e11016] [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: 02/18/2022] [Revised: 04/29/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022] Open
Abstract
Pulmonary arterial remodeling at an early stage, including excessive proliferation and migration of smooth muscle cells, is a hallmark of pulmonary arterial hypertension (PAH). Salt-inducible kinases (SIKs) have been increasingly reported to play a key role in smooth muscle cell proliferation and phenotype switching, which may be associated with arterial remodeling. However, the potential effects of SIK1 in PAH and the underlying mechanisms have not been explored. The aim of this study was to determine whether reduced expression or inactivation of SIK1 is associated with pulmonary arterial remodeling in PAH and to elucidate whether it is related to the Hippo/Yes-associated protein (YAP) pathway. Using mouse models of PAH and hypoxia-stimulated hPASMCs, we observed that SIK1 expression was robustly reduced in lung tissues of PAH mice and hPASMCs cultured under hypoxia. In hypoxia-induced PAH mice, pharmacological SIK inhibition or AAV9-mediated specific smooth muscle SIK1 knockdown strongly aggravated pathological changes caused by hypoxia, including right ventricular hypertrophy and small pulmonary arterial remodeling. Meanwhile, in hypoxia-stimulated hPASMCs, SIK1 knockdown or inhibition promoted proliferation and migration under hypoxia, accompanied by decreased phosphorylation and increased nuclear accumulation of YAP, while SIK1 overexpression inhibited hypoxia-induced proliferation, migration and nuclear translocation of YAP in hPASMCs. YAP knockdown attenuated the increase in cell proliferation induced by HG-9-91-01 treatment or SIK1 siRNA transfection under hypoxia in hPASMCs. Here, we identified SIK1 as an antiproliferative factor in hypoxia-induced pulmonary arterial remodeling via YAP-mediated mechanisms. These results show that targeting SIK1 may be a promising therapeutic strategy for the treatment of PAH.
Collapse
|
13
|
Zhong H, Yu H, Chen J, Mok SWF, Tan X, Zhao B, He S, Lan L, Fu X, Chen G, Zhu D. The short-chain fatty acid butyrate accelerates vascular calcification via regulation of histone deacetylases and NF-κB signaling. Vascul Pharmacol 2022; 146:107096. [PMID: 35952961 DOI: 10.1016/j.vph.2022.107096] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022]
Abstract
Recent studies have shown that short-chain fatty acids (SCFAs), primarily acetate, propionate and butyrate, play a crucial role in the pathogenesis of cardiovascular disease. Whether SCFAs regulate vascular calcification, a common pathological change in cardiovascular tissues, remains unclear. This study aimed to investigate the potential role of SCFAs in vascular calcification. Using cellular and animal models of vascular calcification, we showed that butyrate significantly enhanced high phosphate (Pi)-induced calcification and osteogenic transition of vascular smooth muscle cells (VSMC) in vitro, whereas acetate and propionate had no effects. Subsequent studies confirmed that butyrate significantly promoted high Pi-induced aortic ring calcification ex vivo and high dose vitamin D3 (vD3)-induced mouse vascular calcification in vivo. Mechanistically, butyrate significantly inhibited histone deacetylase (HDAC) expression in VSMCs, and a pan HDAC inhibitor Trichostatin A showed similar inductive effects on calcification and osteogenic transition of VSMCs to butyrate. In addition, the SCFA sensing receptors Gpr41 and Gpr109a were primarily expressed by VSMCs, and butyrate induced the rapid activation of NF-κB, Wnt and Akt signaling in VSMCs. Intriguingly, the NF-κB inhibitor SC75741 significantly attenuated butyrate-induced calcification and the osteogenic gene Msx2 expression in VSMCs. We showed that knockdown of Gpr41 but not Gpr109a attenuated butyrate-induced VSMC calcification. This study reveals that butyrate accelerates vascular calcification via its dual effects on HDAC inhibition and NF-κB activation. Our data provide novel insights into the role of microbe-host interaction in vascular calcification, and may have implications for the development of potential therapy for vascular calcification.
Collapse
Affiliation(s)
- Hui Zhong
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Hongjiao Yu
- Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Jiaxin Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Simon Wing Fai Mok
- Faculty of Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Xiao Tan
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Bohou Zhao
- Emergency Department, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shengping He
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lan Lan
- Department of Anesthesiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| | - Guojun Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, Guangdong 510260, China; Department of Biochemistry and Molecular Biology, GMU-GIBH Joint School of Life Science, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
14
|
Lan Z, Chen A, Li L, Ye Y, Liang Q, Dong Q, Wang S, Fu M, Li Y, Liu X, Zhu Z, Ou JS, Qiu X, Lu L, Yan J. Downregulation of HDAC9 by the ketone metabolite β-hydroxybutyrate suppresses vascular calcification. J Pathol 2022; 258:213-226. [PMID: 35894849 DOI: 10.1002/path.5992] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/17/2022] [Accepted: 07/23/2022] [Indexed: 11/07/2022]
Abstract
Vascular calcification is an actively regulated process resembling bone formation and contributes to the cardiovascular morbidity and mortality of chronic kidney disease (CKD). However, effective therapy for vascular calcification is still lacking. The ketone body β-hydroxybutyrate (BHB) has been demonstrated to have health-promoting effects including anti-inflammation and cardiovascular protective effects. However, whether BHB protects against vascular calcification in CKD remains unclear. In this study, Alizarin Red staining and calcium content assay showed that BHB reduced calcification of vascular smooth muscle cells (VSMCs) and arterial rings. Of note, compared with CKD patients without thoracic calcification, serum BHB levels were lower in CKD patients with thoracic calcification. Supplementation with 1,3-butanediol (1,3-B), the precursor of BHB, attenuated aortic calcification in CKD rats and VitD3-overloaded mice. Furthermore, RNA-Seq analysis revealed that BHB downregulated HDAC9, which was further confirmed by RT-qPCR and western blot analysis. Both pharmacological inhibition and knockdown of HDAC9 attenuated calcification of human VSMCs, while overexpression of HDAC9 exacerbated calcification of VSMCs and aortic rings, indicating that HDAC9 promotes vascular calcification under CKD conditions. Of note, BHB treatment antagonized HDAC9-induced vascular calcification. In addition, HDAC9 overexpression activated NF-κB signaling pathway and inhibition of NF-κB attenuated HDAC9-induced VSMC calcification, suggesting that HDAC9 promotes vascular calcification via activation of NF-κB. In conclusion, our study demonstrates that BHB supplementation inhibits vascular calcification in CKD via modulation of the HDAC9-dependent NF-κB signaling pathway. Moreover, we unveil a crucial mechanistic role of HDAC9 in vascular calcification under CKD conditions, thus nutritional intervention or pharmacological approaches to enhance BHB levels could act as promising therapeutic strategies to target HDAC9 for the treatment of vascular calcification in CKD. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Zirong Lan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| | - An Chen
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| | - Li Li
- Department of Cardiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, PR China
| | - Yuanzhi Ye
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| | - Qingchun Liang
- Department of Anesthesiology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, PR China
| | - Qianqian Dong
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| | - Siyi Wang
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| | - Mingwei Fu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| | - Yining Li
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| | - Xiaoyu Liu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| | - Zhenyu Zhu
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| | - Jing-Song Ou
- Division of Cardiac Surgery, National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, NHC key Laboratory of Assisted Circulation, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, PR China
| | - Xiaozhong Qiu
- The Fifth Affiliated Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering; School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Lihe Lu
- Department of Pathophysiology, Zhongshan Medical School, Sun Yat-Sen University, Guangzhou, PR China
| | - Jianyun Yan
- Department of Cardiology, Laboratory of Heart Center, Heart Center, Zhujiang Hospital, Southern Medical University; Guangdong Provincial Key Laboratory of Shock and Microcirculation; Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease; Sino-Japanese Cooperation Platform for Translational Research in Heart Failure, Guangzhou, PR China
| |
Collapse
|
15
|
Epigenetic regulation in cardiovascular disease: mechanisms and advances in clinical trials. Signal Transduct Target Ther 2022; 7:200. [PMID: 35752619 PMCID: PMC9233709 DOI: 10.1038/s41392-022-01055-2] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/18/2022] [Accepted: 06/08/2022] [Indexed: 12/17/2022] Open
Abstract
Epigenetics is closely related to cardiovascular diseases. Genome-wide linkage and association analyses and candidate gene approaches illustrate the multigenic complexity of cardiovascular disease. Several epigenetic mechanisms, such as DNA methylation, histone modification, and noncoding RNA, which are of importance for cardiovascular disease development and regression. Targeting epigenetic key enzymes, especially the DNA methyltransferases, histone methyltransferases, histone acetylases, histone deacetylases and their regulated target genes, could represent an attractive new route for the diagnosis and treatment of cardiovascular diseases. Herein, we summarize the knowledge on epigenetic history and essential regulatory mechanisms in cardiovascular diseases. Furthermore, we discuss the preclinical studies and drugs that are targeted these epigenetic key enzymes for cardiovascular diseases therapy. Finally, we conclude the clinical trials that are going to target some of these processes.
Collapse
|
16
|
van Andel MM, Groenink M, van den Berg MP, Timmermans J, Scholte AJHA, Mulder BJM, Zwinderman AH, de Waard V. Genome-wide methylation patterns in Marfan syndrome. Clin Epigenetics 2021; 13:217. [PMID: 34895303 PMCID: PMC8665617 DOI: 10.1186/s13148-021-01204-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/27/2021] [Indexed: 12/29/2022] Open
Abstract
Background Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the Fibrillin-1 gene (FBN1). Here, we undertook the first epigenome-wide association study (EWAS) in patients with MFS aiming at identifying DNA methylation loci associated with MFS phenotypes that may shed light on the disease process. Methods The Illumina 450 k DNA-methylation array was used on stored peripheral whole-blood samples of 190 patients with MFS originally included in the COMPARE trial. An unbiased genome-wide approach was used, and methylation of CpG-sites across the entire genome was evaluated. Additionally, we investigated CpG-sites across the FBN1-locus (15q21.1) more closely, since this is the gene defective in MFS. Differentially Methylated Positions (DMPs) and Differentially Methylated Regions (DMRs) were identified through regression analysis. Associations between methylation levels and aortic diameters and presence or absence of 21 clinical features of MFS at baseline were analyzed. Moreover, associations between aortic diameter change, and the occurrence of clinical events (death any cause, type-A or -B dissection/rupture, or aortic surgery) and methylation levels were analyzed. Results We identified 28 DMPs that are significantly associated with aortic diameters in patients with MFS. Seven of these DMPs (25%) could be allocated to a gene that was previously associated with cardiovascular diseases (HDAC4, IGF2BP3, CASZ1, SDK1, PCDHGA1, DIO3, PTPRN2). Moreover, we identified seven DMPs that were significantly associated with aortic diameter change and five DMP’s that associated with clinical events. No significant associations at p < 10–8 or p < 10–6 were found with any of the non-cardiovascular phenotypic MFS features. Investigating DMRs, clusters were seen mostly on X- and Y, and chromosome 18–22. The remaining DMRs indicated involvement of a large family of protocadherins on chromosome 5, which were not reported in MFS before. Conclusion This EWAS in patients with MFS has identified a number of methylation loci significantly associated with aortic diameters, aortic dilatation rate and aortic events. Our findings add to the slowly growing literature on the regulation of gene expression in MFS patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01204-4.
Collapse
Affiliation(s)
- Mitzi M van Andel
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| | - Maarten Groenink
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.,Department of Radiology, Amsterdam UMC, Amsterdam, The Netherlands
| | - Maarten P van den Berg
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Janneke Timmermans
- Department of Cardiology, Radboud University Hospital, Nijmegen, The Netherlands
| | - Arthur J H A Scholte
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Barbara J M Mulder
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Aeilko H Zwinderman
- Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Vivian de Waard
- Department of Medical Biochemistry, Amsterdam UMC, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| |
Collapse
|
17
|
Cai Y, Wang XL, Lu J, Lin X, Dong J, Guzman RJ. Salt-Inducible Kinase 3 Promotes Vascular Smooth Muscle Cell Proliferation and Arterial Restenosis by Regulating AKT and PKA-CREB Signaling. Arterioscler Thromb Vasc Biol 2021; 41:2431-2451. [PMID: 34196217 PMCID: PMC8411910 DOI: 10.1161/atvbaha.121.316219] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 06/17/2021] [Indexed: 01/11/2023]
Abstract
Objective Arterial restenosis is the pathological narrowing of arteries after endovascular procedures, and it is an adverse event that causes patients to experience recurrent occlusive symptoms. Following angioplasty, vascular smooth muscle cells (SMCs) change their phenotype, migrate, and proliferate, resulting in neointima formation, a hallmark of arterial restenosis. SIKs (salt-inducible kinases) are a subfamily of the AMP-activated protein kinase family that play a critical role in metabolic diseases including hepatic lipogenesis and glucose metabolism. Their role in vascular pathological remodeling, however, has not been explored. In this study, we aimed to understand the role and regulation of SIK3 in vascular SMC migration, proliferation, and neointima formation. Approach and Results We observed that SIK3 expression was low in contractile aortic SMCs but high in proliferating SMCs. It was also highly induced by growth medium in vitro and in neointimal lesions in vivo. Inactivation of SIKs significantly attenuated vascular SMC proliferation and up-regulated p21CIP1 and p27KIP1. SIK inhibition also suppressed SMC migration and modulated actin polymerization. Importantly, we found that inhibition of SIKs reduced neointima formation and vascular inflammation in a femoral artery wire injury model. In mechanistic studies, we demonstrated that inactivation of SIKs mainly suppressed SMC proliferation by down-regulating AKT (protein kinase B) and PKA (protein kinase A)-CREB (cAMP response element-binding protein) signaling. CRTC3 (CREB-regulated transcriptional coactivator 3) signaling likely contributed to SIK inactivation-mediated antiproliferative effects. Conclusions These findings suggest that SIK3 may play a critical role in regulating SMC proliferation, migration, and arterial restenosis. This study provides insights into SIK inhibition as a potential therapeutic strategy for treating restenosis in patients with peripheral arterial disease.
Collapse
MESH Headings
- Animals
- CREB-Binding Protein/metabolism
- Cell Movement
- Cell Proliferation/drug effects
- Cells, Cultured
- Constriction, Pathologic
- Cyclic AMP-Dependent Protein Kinases/metabolism
- Cyclin-Dependent Kinase Inhibitor p21/genetics
- Cyclin-Dependent Kinase Inhibitor p21/metabolism
- Cyclin-Dependent Kinase Inhibitor p27/genetics
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Disease Models, Animal
- Female
- Femoral Artery/enzymology
- Femoral Artery/injuries
- Femoral Artery/pathology
- Male
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/injuries
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/pathology
- Neointima
- Phenylurea Compounds/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins c-akt/metabolism
- Pyrimidines/pharmacology
- Rats, Sprague-Dawley
- Signal Transduction
- Transcription Factors/genetics
- Transcription Factors/metabolism
- Vascular System Injuries/drug therapy
- Vascular System Injuries/enzymology
- Vascular System Injuries/genetics
- Vascular System Injuries/pathology
- Mice
- Rats
Collapse
Affiliation(s)
- Yujun Cai
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale University School of Medicine, New Haven, CT 06510
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Xue-Lin Wang
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Jinny Lu
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Xin Lin
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115
| | - Jonathan Dong
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Raul J Guzman
- Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Yale University School of Medicine, New Haven, CT 06510
- Division of Vascular and Endovascular Surgery, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
18
|
Shkedi O, Kehat I. SIKs (Salt-Inducible Kinases) in Arterial Restenosis. Arterioscler Thromb Vasc Biol 2021; 41:2452-2453. [PMID: 34320836 DOI: 10.1161/atvbaha.121.316691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Omer Shkedi
- The Department of Physiology, Biophysics and System Biology, The Rapport Institute and the Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa (O.S., I.K.)
| | - Izhak Kehat
- The Department of Physiology, Biophysics and System Biology, The Rapport Institute and the Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa (O.S., I.K.).,Department of Cardiology and the Clinical Research Institute at Rambam, Rambam Medical Center, Haifa, Israel (I.K.)
| |
Collapse
|
19
|
Badawi M, Mori T, Kurihara T, Yoshizawa T, Nohara K, Kouyama-Suzuki E, Yanagawa T, Shirai Y, Tabuchi K. Risperidone Mitigates Enhanced Excitatory Neuronal Function and Repetitive Behavior Caused by an ASD-Associated Mutation of SIK1. Front Mol Neurosci 2021; 14:706494. [PMID: 34295222 PMCID: PMC8289890 DOI: 10.3389/fnmol.2021.706494] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 05/27/2021] [Indexed: 12/28/2022] Open
Abstract
Six mutations in the salt-inducible kinase 1 (SIK1)-coding gene have been identified in patients with early infantile epileptic encephalopathy (EIEE-30) accompanied by autistic symptoms. Two of the mutations are non-sense mutations that truncate the C-terminal region of SIK1. It has been shown that the C-terminal-truncated form of SIK1 protein affects the subcellular distribution of SIK1 protein, tempting to speculate the relevance to the pathophysiology of the disorders. We generated SIK1-mutant (SIK1-MT) mice recapitulating the C-terminal-truncated mutations using CRISPR/Cas9-mediated genome editing. SIK1-MT protein was distributed in the nucleus and cytoplasm, whereas the distribution of wild-type SIK1 was restricted to the nucleus. We found the disruption of excitatory and inhibitory (E/I) synaptic balance due to an increase in excitatory synaptic transmission and enhancement of neural excitability in the pyramidal neurons in layer 5 of the medial prefrontal cortex in SIK1-MT mice. We also found the increased repetitive behavior and social behavioral deficits in SIK1-MT mice. The risperidone administration attenuated the neural excitability and excitatory synaptic transmission, but the disrupted E/I synaptic balance was unchanged, because it also reduced the inhibitory synaptic transmission. Risperidone also eliminated the repetitive behavior but not social behavioral deficits. These results indicate that risperidone has a role in decreasing neuronal excitability and excitatory synapses, ameliorating repetitive behavior in the SIK1-truncated mice.
Collapse
Affiliation(s)
- Moataz Badawi
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takuma Mori
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto, Japan.,Department of NeuroHealth Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Taiga Kurihara
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Takahiro Yoshizawa
- Research Center for Supports to Advanced Science, Shinshu University, Matsumoto, Japan
| | - Katsuhiro Nohara
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Emi Kouyama-Suzuki
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Toru Yanagawa
- Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yoshinori Shirai
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Katsuhiko Tabuchi
- Department of Molecular and Cellular Physiology, Shinshu University School of Medicine, Matsumoto, Japan.,Department of NeuroHealth Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| |
Collapse
|
20
|
Nuts and bolts of the salt-inducible kinases (SIKs). Biochem J 2021; 478:1377-1397. [PMID: 33861845 PMCID: PMC8057676 DOI: 10.1042/bcj20200502] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/24/2022]
Abstract
The salt-inducible kinases, SIK1, SIK2 and SIK3, most closely resemble the AMP-activated protein kinase (AMPK) and other AMPK-related kinases, and like these family members they require phosphorylation by LKB1 to be catalytically active. However, unlike other AMPK-related kinases they are phosphorylated by cyclic AMP-dependent protein kinase (PKA), which promotes their binding to 14-3-3 proteins and inactivation. The most well-established substrates of the SIKs are the CREB-regulated transcriptional co-activators (CRTCs), and the Class 2a histone deacetylases (HDAC4/5/7/9). Phosphorylation by SIKs promotes the translocation of CRTCs and Class 2a HDACs to the cytoplasm and their binding to 14-3-3s, preventing them from regulating their nuclear binding partners, the transcription factors CREB and MEF2. This process is reversed by PKA-dependent inactivation of the SIKs leading to dephosphorylation of CRTCs and Class 2a HDACs and their re-entry into the nucleus. Through the reversible regulation of these substrates and others that have not yet been identified, the SIKs regulate many physiological processes ranging from innate immunity, circadian rhythms and bone formation, to skin pigmentation and metabolism. This review summarises current knowledge of the SIKs and the evidence underpinning these findings, and discusses the therapeutic potential of SIK inhibitors for the treatment of disease.
Collapse
|
21
|
Travers JG, Hu T, McKinsey TA. The black sheep of class IIa: HDAC7 SIKens the heart. J Clin Invest 2021; 130:2811-2813. [PMID: 32364532 DOI: 10.1172/jci137074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Class IIa histone deacetylases (HDACs) repress cardiomyocyte hypertrophy through association with the prohypertrophic transcription factor (TF) myocyte enhancer factor-2 (MEF2). The four class IIa HDACs - HDAC4, -5, -7, and -9 - are subject to signal-dependent phosphorylation by members of the Ca2+/calmodulin-dependent protein kinase (CaMK) group. In response to stress, HDAC4, HDAC5, and HDAC9 undergo phosphorylation-induced nuclear export in cardiomyocytes, freeing MEF2 to stimulate progrowth genes; it was generally assumed that HDAC7 is also antihypertrophic. However, in this issue of the JCI, Hsu and colleagues demonstrate that, in sharp contrast to the other class IIa HDACs, HDAC7 is constitutively localized to the cardiomyocyte cytoplasm, where it promotes cardiac hypertrophy. Phosphorylation of HDAC7 by the CaMK group member salt-inducible kinase 1 (SIK1) stabilized the deacetylase, leading to increased expression of c-Myc, which in turn stimulated a pathological gene program. These unexpected findings highlight the SIK1/HDAC7 signaling axis as a promising target for the treatment of cardiac hypertrophy and heart failure.
Collapse
|
22
|
Liu Y, Tang W, Ji C, Gu J, Chen Y, Huang J, Zhao X, Sun Y, Wang C, Guan W, Liu J, Jiang B. The Selective SIK2 Inhibitor ARN-3236 Produces Strong Antidepressant-Like Efficacy in Mice via the Hippocampal CRTC1-CREB-BDNF Pathway. Front Pharmacol 2021; 11:624429. [PMID: 33519490 PMCID: PMC7840484 DOI: 10.3389/fphar.2020.624429] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/04/2020] [Indexed: 12/20/2022] Open
Abstract
Depression is a widespread chronic medical illness affecting thoughts, mood, and physical health. However, the limited and delayed therapeutic efficacy of monoaminergic drugs has led to intensive research efforts to develop novel antidepressants. ARN-3236 is the first potent and selective inhibitor of salt-inducible kinase 2 (SIK2). In this study, a multidisciplinary approach was used to explore the antidepressant-like actions of ARN-3236 in mice. Chronic social defeat stress (CSDS) and chronic unpredictable mild stress (CUMS) models of depression, various behavioral tests, high performance liquid chromatography-tandem mass spectrometry, stereotactic infusion, viral-mediated gene transfer, western blotting, co-immunoprecipitation and immunofluorescence were used together. It was found that ARN-3236 could penetrate the blood-brain barrier. Repeated ARN-3236 administration induced significant antidepressant-like effects in both the CSDS and CUMS models of depression, accompanied with fully preventing the stress-enhanced SIK2 expression and cytoplasmic translocation of cyclic adenosine monophosphate response element binding protein (CREB)-regulated transcription coactivator 1 (CRTC1) in the hippocampus. ARN-3236 treatment also completely reversed the down-regulating effects of CSDS and CUMS on the hippocampal brain-derived neurotrophic factor (BDNF) system and neurogenesis. Moreover, we demonstrated that the hippocampal CRTC1-CREB-BDNF pathway mediated the antidepressant-like efficacy of ARN-3236. Collectively, ARN-3236 possesses strong protecting effects against chronic stress, and could be a novel antidepressant beyond monoaminergic drugs.
Collapse
Affiliation(s)
- Yue Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Wenqian Tang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Chunhui Ji
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Jianghong Gu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Yanmei Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Jie Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Xinyi Zhao
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Yingfang Sun
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Chengniu Wang
- Basic Medical Research Centre, Medical College, Nantong University, Nantong, China
| | - Wei Guan
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, China
| | - Jianfeng Liu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China.,Provincial Key Laboratory of Inflammation and Molecular Drug Target, Nantong, China
| |
Collapse
|
23
|
Chen X, He Y, Fu W, Sahebkar A, Tan Y, Xu S, Li H. Histone Deacetylases (HDACs) and Atherosclerosis: A Mechanistic and Pharmacological Review. Front Cell Dev Biol 2020; 8:581015. [PMID: 33282862 PMCID: PMC7688915 DOI: 10.3389/fcell.2020.581015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis (AS), the most common underlying pathology for coronary artery disease, is a chronic inflammatory, proliferative disease in large- and medium-sized arteries. The vascular endothelium is important for maintaining vascular health. Endothelial dysfunction is a critical early event leading to AS, which is a major risk factor for stroke and myocardial infarction. Accumulating evidence has suggested the critical roles of histone deacetylases (HDACs) in regulating vascular cell homeostasis and AS. The purpose of this review is to present an updated view on the roles of HDACs (Class I, Class II, Class IV) and HDAC inhibitors in vascular dysfunction and AS. We also elaborate on the novel therapeutic targets and agents in atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaona Chen
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanhong He
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenjun Fu
- The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Polish Mother's Memorial Hospital Research Institute, Łódź, Poland
| | - Yuhui Tan
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Suowen Xu
- Department of Endocrinology, First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hong Li
- Department of Medical Biotechnology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,The Research Center of Basic Integrative Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
24
|
Choe N, Shin S, Joung H, Ryu J, Kim Y, Ahn Y, Kook H, Kwon D. The microRNA miR-134-5p induces calcium deposition by inhibiting histone deacetylase 5 in vascular smooth muscle cells. J Cell Mol Med 2020; 24:10542-10550. [PMID: 32783377 PMCID: PMC7521311 DOI: 10.1111/jcmm.15670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 12/30/2022] Open
Abstract
Calcium deposition in vascular smooth muscle cells (VSMCs) is a form of ectopic ossification in blood vessels. It can result in rigidity of the vasculature and an increase in cardiac events. Here, we report that the microRNA miR-134-5p potentiates inorganic phosphate (Pi)-induced calcium deposition in VSMCs by inhibiting histone deacetylase 5 (HDAC5). Using miRNA microarray analysis of Pi-treated rat VSMCs, we first selected miR-134-5p for further evaluation. Quantitative RT-PCR confirmed that miR-134-5p was increased in Pi-treated A10 cells, a rat VSMC line. Transfection of miR-134-5p mimic potentiated the Pi-induced increase in calcium contents. miR-134-5p increased the amounts of bone runt-related transcription factor 2 (RUNX2) protein and bone morphogenic protein 2 (BMP2) mRNA in the presence of Pi but decreased the expression of osteoprotegerin (OPG). Bioinformatic analysis showed that the HDAC5 3'untranslated region (3'UTR) was one of the targets of miR-134-5p. The luciferase construct containing the 3'UTR of HDAC5 was down-regulated by miR-134-5p mimic in a dose-dependent manner in VSMCs. Overexpression of HDAC5 mitigated the calcium deposition induced by miR-134-5p. Our results suggest that a Pi-induced increase of miR-134-5p may cause vascular calcification through repression of HDAC5.
Collapse
Affiliation(s)
- Nakwon Choe
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Sera Shin
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Hosouk Joung
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Juhee Ryu
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Young‐Kook Kim
- Department of BiochemistryChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Youngkeun Ahn
- Department of CardiologyChonnam National University HospitalGwangjuRepublic of Korea
| | - Hyun Kook
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| | - Duk‐Hwa Kwon
- Department of PharmacologyChonnam National University Medical SchoolHwasunRepublic of Korea
| |
Collapse
|
25
|
He P, Yu H, Jiang L, Chen Z, Wang S, Macrae VE, Fu X, Zhu D. Hdac9 inhibits medial artery calcification through down-regulation of Osterix. Vascul Pharmacol 2020; 132:106775. [PMID: 32702412 DOI: 10.1016/j.vph.2020.106775] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/15/2020] [Accepted: 07/15/2020] [Indexed: 02/03/2023]
Abstract
BACKGROUNDS Medial artery calcification (MAC) significantly contributes to the increased cardiovascular death in patients with chronic kidney disease (CKD). Previous genome-wide association studies have shown that various genetic variants of the histone deacetylase Hdac9 are associated with cardiovascular disease, but the role of Hdac9 in MAC under CKD conditions remains unclear. METHODS High phosphate-induced vascular smooth muscle cell (VSMC) calcification and MAC in mice administered with vitamin D3 (vD) were used in the present study. Alizarin red staining, calcium quantitative assay, qPCR, western blotting and histology were performed. RESULTS Hdac9 expression was significantly down-regulated during high phosphate-induced vascular smooth muscle cell (VSMC) calcification and MAC in mice administered with vitamin D3 (vD). Furthermore, high phosphate treatment inhibited phosphorylation of Akt, and pharmacological inhibition of Akt signaling reduced Hdac9 expression in cultured VSMCs. Knockdown of Hdac9 significantly enhanced calcium deposition in VSMCs. Conversely, adenovirus mediated-overexpression of Hdac9 inhibited high phosphate induced VSMC in vitro calcification. Our subsequent mechanistic studies revealed that the anti-calcific effect of Hdac9 was mediated through down-regulation of osteoblast-specific transcription factor Osterix. CONCLUSION These data suggest that Hdac9 is a novel inhibitor of MAC and may represent a potential therapeutic target for MAC in CKD patients.
Collapse
Affiliation(s)
- Pengcheng He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Hongjiao Yu
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Forester hill, Aberdeen AB25 2ZD, UK
| | - Lei Jiang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Ziying Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Siying Wang
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China
| | - Vicky E Macrae
- The Roslin Institute, RDSVS, Easter Bush Campus, University of Edinburgh, Midlothian EH25 9RG, UK
| | - Xiaodong Fu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| | - Dongxing Zhu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510260, China.
| |
Collapse
|
26
|
Roles of Histone Acetylation Modifiers and Other Epigenetic Regulators in Vascular Calcification. Int J Mol Sci 2020; 21:ijms21093246. [PMID: 32375326 PMCID: PMC7247359 DOI: 10.3390/ijms21093246] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/28/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification (VC) is characterized by calcium deposition inside arteries and is closely associated with the morbidity and mortality of atherosclerosis, chronic kidney disease, diabetes, and other cardiovascular diseases (CVDs). VC is now widely known to be an active process occurring in vascular smooth muscle cells (VSMCs) involving multiple mechanisms and factors. These mechanisms share features with the process of bone formation, since the phenotype switching from the contractile to the osteochondrogenic phenotype also occurs in VSMCs during VC. In addition, VC can be regulated by epigenetic factors, including DNA methylation, histone modification, and noncoding RNAs. Although VC is commonly observed in patients with chronic kidney disease and CVD, specific drugs for VC have not been developed. Thus, discovering novel therapeutic targets may be necessary. In this review, we summarize the current experimental evidence regarding the role of epigenetic regulators including histone deacetylases and propose the therapeutic implication of these regulators in the treatment of VC.
Collapse
|
27
|
The Epigenetic Landscape of Vascular Calcification: An Integrative Perspective. Int J Mol Sci 2020; 21:ijms21030980. [PMID: 32024140 PMCID: PMC7037112 DOI: 10.3390/ijms21030980] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/14/2020] [Accepted: 01/15/2020] [Indexed: 12/26/2022] Open
Abstract
Vascular calcification (VC) is an important complication among patients of advanced age, those with chronic kidney disease, and those with diabetes mellitus. The pathophysiology of VC encompasses passive occurrence of physico-chemical calcium deposition, active cellular secretion of osteoid matrix upon exposure to metabolically noxious stimuli, or a variable combination of both processes. Epigenetic alterations have been shown to participate in this complex environment, through mechanisms including DNA methylation, non-coding RNAs, histone modifications, and chromatin changes. Despite such importance, existing reviews fail to provide a comprehensive view of all relevant reports addressing epigenetic processes in VC, and cross-talk between different epigenetic machineries is rarely examined. We conducted a systematic review based on PUBMED and MEDLINE databases up to 30 September 2019, to identify clinical, translational, and experimental reports addressing epigenetic processes in VC; we retrieved 66 original studies, among which 60.6% looked into the pathogenic role of non-coding RNA, followed by DNA methylation (12.1%), histone modification (9.1%), and chromatin changes (4.5%). Nine (13.6%) reports examined the discrepancy of epigenetic signatures between subjects or tissues with and without VC, supporting their applicability as biomarkers. Assisted by bioinformatic analyses blending in each epigenetic component, we discovered prominent interactions between microRNAs, DNA methylation, and histone modification regarding potential influences on VC risk.
Collapse
|
28
|
Jiang B, Wang H, Wang JL, Wang YJ, Zhu Q, Wang CN, Song L, Gao TT, Wang Y, Meng GL, Wu F, Ling Y, Zhang W, Li JX. Hippocampal Salt-Inducible Kinase 2 Plays a Role in Depression via the CREB-Regulated Transcription Coactivator 1-cAMP Response Element Binding-Brain-Derived Neurotrophic Factor Pathway. Biol Psychiatry 2019; 85:650-666. [PMID: 30503507 DOI: 10.1016/j.biopsych.2018.10.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Developing novel pharmacological targets beyond monoaminergic systems is now a popular strategy for finding new ways to treat depression. Salt-inducible kinase (SIK) is a kinase that regulates the nuclear translocation of cyclic adenosine monophosphate response element binding protein (CREB)-regulated transcription coactivator (CRTC) by phosphorylation. Here, we hypothesize that dysfunction of the central SIK-CRTC system may contribute to the pathogenesis of depression. METHODS Chronic social defeat stress (CSDS) and chronic unpredictable mild stress (CUMS) models of depression, various behavioral tests, viral-mediated gene transfer, Western blotting, coimmunoprecipitation, quantitative real-time reverse transcription polymerase chain reaction, and immunohistochemistry were used in this study (for in vivo studies, n = 10; for in vitro studies, n = 5). RESULTS Both CSDS and CUMS markedly increased the expression of hippocampal SIK2, which reduced CRTC1 nuclear translocation and binding of CRTC1 and CREB in the hippocampus. Genetic overexpression of hippocampal SIK2 in naïve mice simulated chronic stress, inducing depressive-like behaviors in the forced swim test, tail suspension test, sucrose preference test, and social interaction test, as well as decreasing the brain-derived neurotrophic factor signaling cascade and neurogenesis in the hippocampus. In contrast, genetic knockdown and knockout of hippocampal SIK2 protected against CSDS and CUMS, exerting significant antidepressant-like effects that were mediated via the downstream CRTC1-CREB-brain-derived neurotrophic factor pathway. Moreover, fluoxetine, venlafaxine, and mirtazapine all significantly restored the effects of CSDS and CUMS on the hippocampal SIK2-CRTC1 pathway, which was necessary for their antidepressant actions. CONCLUSIONS The hippocampal SIK2-CRTC1 pathway is involved in the pathogenesis of depression, and hippocampal SIK2 could be a novel target for the development of antidepressants.
Collapse
Affiliation(s)
- Bo Jiang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China.
| | - Hao Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Jin-Liang Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Ying-Jie Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Qing Zhu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Cheng-Niu Wang
- Basic Medical Research Centre, Medical College, Nantong University, Nantong, Jiangsu, China
| | - Lu Song
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Ting-Ting Gao
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Yuan Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Guo-Liang Meng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Feng Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Yong Ling
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Wei Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China; Jiangsu Province Key Laboratory of Inflammation and Molecular Drug Target, Nantong, Jiangsu, China
| | - Jun-Xu Li
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
29
|
Abend A, Shkedi O, Fertouk M, Caspi LH, Kehat I. Salt-inducible kinase induces cytoplasmic histone deacetylase 4 to promote vascular calcification. EMBO Rep 2017; 18:1166-1185. [PMID: 28588072 PMCID: PMC5494505 DOI: 10.15252/embr.201643686] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 04/18/2017] [Accepted: 04/26/2017] [Indexed: 12/25/2022] Open
Abstract
A pathologic osteochondrogenic differentiation of vascular smooth muscle cells (VSMCs) promotes arterial calcifications, a process associated with significant morbidity and mortality. The molecular pathways promoting this pathology are not completely understood. We studied VSMCs, mouse aortic rings, and human aortic valves and showed here that histone deacetylase 4 (HDAC4) is upregulated early in the calcification process. Gain- and loss-of-function assays demonstrate that HDAC4 is a positive regulator driving this pathology. HDAC4 can shuttle between the nucleus and cytoplasm, but in VSMCs, the cytoplasmic rather than the nuclear activity of HDAC4 promotes calcification, and a nuclear-localized mutant of HDAC4 fails to promote calcification. The cytoplasmic location and function of HDAC4 is controlled by the activity of salt-inducible kinase (SIK). Pharmacologic inhibition of SIK sends HDAC4 to the nucleus and inhibits the calcification process in VSMCs, aortic rings, and in vivo In the cytoplasm, HDAC4 binds and its activity depends on the adaptor protein ENIGMA (Pdlim7) to promote vascular calcification. These results establish a cytoplasmic role for HDAC4 and identify HDAC4, SIK, and ENIGMA as mediators of vascular calcification.
Collapse
Affiliation(s)
- Alon Abend
- The Rappaport Institute and the Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Omer Shkedi
- The Rappaport Institute and the Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Michal Fertouk
- The Rappaport Institute and the Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Department of Cardiac Surgery, Rambam Medical Center, Haifa, Israel
| | - Lilac H Caspi
- The Rappaport Institute and the Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Izhak Kehat
- The Rappaport Institute and the Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Department of Cardiology and the Clinical Research Institute at Rambam, Rambam Medical Center, Haifa, Israel
| |
Collapse
|