1
|
Qi Q, Li L, Liang H, Zeng Y. Role and research progress of histone modification in cardiovascular diseases (Review). Exp Ther Med 2025; 30:132. [PMID: 40421232 PMCID: PMC12105096 DOI: 10.3892/etm.2025.12882] [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: 11/18/2024] [Accepted: 04/11/2025] [Indexed: 05/28/2025] Open
Abstract
As society evolves and lifestyles change, there has been a notable rise in the incidence of cardiovascular diseases due to a parallel rise in associated risk factors. In recent years, considerable research has been conducted on the impact of histone modifications in relation to these conditions. Processes such as acetylation, methylation and phosphorylation of histones, mediated by specific enzymes, are essential in the regulation of gene expression, which in turn influences cellular functions and the progression of diseases. Research shows that alterations in specific histone modifications are closely linked to the onset and advancement of cardiovascular conditions. For instance, significant variations in histone deacetylases and H3K27 methylation have been observed in cases of heart failure and myocardial ischemia-reperfusion injury. In the present review, it was aimed to summarize recent findings in this area, providing a foundation for further exploration of the mechanisms by which histone modifications contribute to cardiovascular diseases.
Collapse
Affiliation(s)
- Qing Qi
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Institute of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Lin Li
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Institute of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Hao Liang
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Institute of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Yidi Zeng
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Institute of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
2
|
Kayzuka C, Rondon-Pereira VC, Nogueira Tavares C, Pacheco Pachado M, Monica FZ, Tanus-Santos JE, Lacchini R. Epigenetics is involved in the pleiotropic effects of statins. Expert Opin Drug Metab Toxicol 2025; 21:689-701. [PMID: 40208655 DOI: 10.1080/17425255.2025.2491732] [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: 08/31/2024] [Revised: 02/18/2025] [Accepted: 04/07/2025] [Indexed: 04/11/2025]
Abstract
INTRODUCTION Statins have significantly reduced mortality from cardiovascular diseases by lowering serum cholesterol levels. Beyond their lipid-lowering effects, statins improve vascular function, reduce inflammation, decrease reactive oxygen species (ROS) formation, and stabilize atherosclerotic plaques. However, the mechanisms underlying these pleiotropic effects remain unclear. AREA COVERED This narrative review summarizes and discusses epigenetic mechanisms that may explain part of the pleiotropic effects of statins. This approach allows for a reevaluation of statin use beyond its cholesterol-lowering benefits. A structured search was conducted in the PubMed and Scopus databases using specific search terms, including articles published up to August 2024. EXPERT OPINION The pleiotropic effects of statins, including those mediated by the isoprenoid pathway, partially explain their clinical benefits. By inhibiting histone deacetylases (HDACs, the 'erasers') and DNA methyltransferases (DNMTs, the 'writers'), statins promote increased histone acetylation and reduced DNA methylation at gene promoter regions. These epigenetic modifications enhance chromatin accessibility, facilitating gene transcription and protecting the cardiovascular system. Further investigation into these epigenetic mechanisms could support the repositioning of statins for broader therapeutic applications. Statins may have benefits extending beyond their role in managing hypercholesterolemia, as their pleiotropic effects contribute to the prevention of cardiovascular disease-related mortality through mechanisms independent of LDL cholesterol reduction.
Collapse
Affiliation(s)
- Cezar Kayzuka
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| | | | - Cecilia Nogueira Tavares
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Mayra Pacheco Pachado
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Fabiola Zakia Monica
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Jose Eduardo Tanus-Santos
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Riccardo Lacchini
- Department of Psychiatric Nursing and Human Sciences, Ribeirao Preto College of Nursing, University of Sao Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
3
|
Li M, Yang L, Wang Y, Zhang L. Comprehensive analysis of diagnostic biomarkers related to histone acetylation in acute myocardial infarction. BMC Med Genomics 2025; 18:75. [PMID: 40251588 DOI: 10.1186/s12920-025-02135-2] [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: 02/27/2024] [Accepted: 03/27/2025] [Indexed: 04/20/2025] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) has become a serious disease that endangers human health, with high morbidity and mortality. Numerous studies have reported histone acetylation can result in the occurrence of cardiovascular diseases. This article aims to explore the potential biomarkers of histone acetylation regulatory genes (ARGs) in AMI patients. METHODS Five AMI datasets were downloaded from the Gene Expression Omnibus (GEO) database. Next, ARG-related genes were gathered by gene set variation analysis (GSVA) and Spearman's correlation analysis. Subsequently, weighted gene co-expression network analysis (WGCNA) was performed to identify the module genes related to histone acetylation regulation. In the GSE60993 and GSE48060 datasets, the common differentially expressed genes (DEGs) between AMI and control samples were screened. Importantly, the intersecting genes were obtained by overlapping ARGs-related genes, common DEGs, and module genes. Then, the biomarkers in AMI were determined by machine learning, receiver operating characteristic (ROC) curves, and quantitative PCR (qPCR). In addition, immune analysis, drug prediction, molecular docking, and the lncRNA-miRNA-mRNA regulatory network targeting the biomarkers were analyzed, respectively. RESULTS Here, a total of 18 intersecting genes were identified by overlapping 7,349 ARGs-related genes, 5,565 module genes, and 25 common DEGs. Further, five biomarkers (AQP9, HLA-DQA1, MCEMP1, NKG7, and S100A12) were obtained, and a nomogram was constructed and verified based on these biomarkers. Notably, the biomarkers were significantly associated with CD8 T cells and neutrophils. In addition, the drugs related to biomarkers were predicted, and ATOGEPANT with the molecular target (S100A12) had a high binding affinity (docking score = -10 kcal/mol). CONCLUSION AQP9, HLA-DQA1, MCEMP1, NKG7, and S100A12 were identified as biomarkers related to ARGs in AMI, which provides a new perspective to study the relationship between ARGs and AMI.
Collapse
Affiliation(s)
- Man Li
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, China
- Tongji Shanxi Hospital, Taiyuan, China
- Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lifeng Yang
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, China
- Tongji Shanxi Hospital, Taiyuan, China
- Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Yan Wang
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, China
- Tongji Shanxi Hospital, Taiyuan, China
- Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Zhang
- Department of Cardiology, Shanxi Bethune Hospital, Taiyuan, China.
- Tongji Shanxi Hospital, Taiyuan, China.
- Third Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
4
|
Manea SA, Vlad ML, Lazar AG, Muresian H, Simionescu M, Manea A. SET7 lysine methyltransferase mediates the up-regulation of NADPH oxidase expression, oxidative stress, and NLRP3 inflammasome priming in atherosclerosis. J Transl Med 2025; 23:339. [PMID: 40098010 PMCID: PMC11912627 DOI: 10.1186/s12967-025-06338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/01/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Dysregulation of histone methylation-based epigenetic mechanisms leads to either transient or long-lasting transcriptomic alterations in vascular and immune cells with important consequences on atherosclerotic plaque development and stability. We hypothesized that the epigenetic enzyme SET7 lysine methyltransferase contributes to the up-regulation of NADPH oxidase (Nox) and NLRP3 inflammasome expression in atherosclerosis. METHODS To test this hypothesis, we examined human non-atherosclerotic and atherosclerotic tissue samples, apolipoprotein E-deficient (ApoE-/-) mice, and human macrophages (Mac) employing real-time PCR, Western blot, immunofluorescence microscopy, and histological techniques. Male ApoE-/- mice with established atherosclerosis were randomized to receive concomitant with the high-fat diet, 5 mg/kg (R)-PFI-2, a selective SET7 pharmacological inhibitor, or its vehicle, every other day for 4 weeks. RESULTS The results revealed that SET7 mRNA and protein, and H3K4me1 levels were significantly elevated in human carotid atherosclerotic lesions, aorta of atherosclerotic mice, and in cultured pro-inflammatory Mac. In the atherosclerotic mice, pharmacological blockade of SET7 catalytic activity with the specific inhibitor, significantly reduced atherosclerotic plaque development, decreased the aortic up-regulation of mRNA and protein levels of Nox catalytic subunits, mitigated the formation of NT-/4HNE-protein adducts, attenuated NLRP3 gene and protein expression, and reduced pro-caspase-1 and pro-IL18 cleavage. In polarized pro-inflammatory human M1-Mac, SET7-oriented pharmacological intervention reduced the transcriptional up-regulation of Nox catalytic subunits, NLRP3, caspase-1, IL1β, and IL18, and the secretion IL1β and TNFα. Transient overexpression of SET7 in human endothelial cells enhanced mRNA levels of Nox1, Nox2, Nox4, Nox5, and p22phox. CONCLUSION The novel results show that SET7 regulates important mechanisms leading to enhanced formation of reactive oxygen species and pro-inflammatory cytokines release in atherosclerosis. The data recommend SET7 as a promising target for pharmacological interventions and as supportive therapeutic strategy in atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Simona-Adriana Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, Bucharest, 050568, Romania
| | - Mihaela-Loredana Vlad
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, Bucharest, 050568, Romania
| | - Alexandra-Gela Lazar
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, Bucharest, 050568, Romania
| | - Horia Muresian
- Cardiovascular Surgery Department, University Hospital Bucharest, Bucharest, Romania
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, Bucharest, 050568, Romania
| | - Adrian Manea
- Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 8, B.P. Hasdeu Street, Bucharest, 050568, Romania.
| |
Collapse
|
5
|
Ji Y, Chen Z, Cai J. Roles and mechanisms of histone methylation in vascular aging and related diseases. Clin Epigenetics 2025; 17:35. [PMID: 39988699 PMCID: PMC11849368 DOI: 10.1186/s13148-025-01842-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 02/14/2025] [Indexed: 02/25/2025] Open
Abstract
The global aging trend has posed significant challenges, rendering healthcare for older adults a crucial focus in medical research. Among the numerous health concerns related to aging, vascular aging and dysfunction are important risk factors and underlying causes of age-related diseases. Histone methylation and demethylation, which are involved in gene expression and cellular senescence, are closely associated with the occurrence and development of vascular aging. Consequently, this review aimed to identify the role of histone methylation in the pathogenesis of vascular aging and its potential for treating age-related vascular diseases and provided new insights into therapeutic strategies targeting the vascular system.
Collapse
Affiliation(s)
- Yufei Ji
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Peking Union Medical College, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhenzhen Chen
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Jun Cai
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Peking Union Medical College, Fuwai Hospital, Chinese Academy of Medical Sciences, Beijing, China.
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Fukumoto T, Shimosawa T, Yakabe M, Yoshida S, Yoshida Y. Recent advances in biomarkers for senescence: Bridging basic research to clinic. Geriatr Gerontol Int 2025; 25:139-147. [PMID: 39754295 DOI: 10.1111/ggi.15054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 10/31/2024] [Accepted: 12/14/2024] [Indexed: 01/06/2025]
Abstract
In this review, we review the current status of biomarkers for aging and possible perspectives on anti-aging or rejuvenation from the standpoint of biomarkers. Aging is observed in all cells and organs, and we focused on research into senescence in the skin, musculoskeletal system, immune system, and cardiovascular system. Commonly used biomarkers include SA-βgal, cell-cycle markers, senescence-associated secretory phenotype (SASP) factors, damage-associated molecular patterns (DAMPs), and DNA-damage-related markers. In addition, each organ or cell has its specific markers. Generally speaking, a combination of biomarkers is required to define age-related changes. When considering the translation of basic research, biomarkers that are highly sensitive, highly specific, with validation and reliability as well as being non-invasive are optimal; however, currently reported markers do not fulfill the prerequisite for biomarkers. In addition, rodent models of aging do not necessarily represent human aging, and markers in rodent or cell models are not applicable in clinical settings. The prerequisite of clinically applicable biomarkers is that they provide useful information for clinical decision-making, such as predicting disease risk, diagnosing disease, monitoring disease progression, or guiding treatment decisions. Therefore, the development of non-invasive robust, reliable, and useful biomarkers in humans is necessary to develop anti-aging therapy for humans. Geriatr Gerontol Int 2025; 25: 139-147.
Collapse
Affiliation(s)
- Takeshi Fukumoto
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tatsuo Shimosawa
- Department of Clinical Laboratory, Graduate School of Medicine, International University of Health and Welfare, Hyogo, Japan
| | - Mitsutaka Yakabe
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shota Yoshida
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yohko Yoshida
- Department of Advanced Senotherapeutics and Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
7
|
Duddu S, Katakia YT, Chakrabarti R, Sharma P, Shukla PC. New epigenome players in the regulation of PCSK9-H3K4me3 and H3K9ac alterations by statin in hypercholesterolemia. J Lipid Res 2025; 66:100699. [PMID: 39566851 PMCID: PMC11699316 DOI: 10.1016/j.jlr.2024.100699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/04/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024] Open
Abstract
Statins are the most effective drugs used worldwide to lower the serum LDL-C by inhibiting the rate-limiting step, HMG-CoA reductase, in cholesterol biosynthesis. Despite its prevalent use, statins are known to increase proprotein convertase subtilisin/kexin type 9 (PCSK9) expression, hindering its efficiency. However, the underlying mechanisms remain elusive. In this study, we have unraveled the pleiotropic effects of statins on hypercholesterolemia via epigenetic regulation of PCSK9. We observed that atorvastatin (ATS) increases the fold enrichment of H3K4me3 at the promoter of PCSK9 by elevating the expression of the SET1/COMPASS family of proteins like SET1b and MLL1 in HepG2. In addition, ATS also acetylates H3K9 by increasing the expression of acetyltransferases like CBP and PCAF. Similarly, in mice fed a high-fat diet, ATS showed increased levels of H3K4me3 and H3K9ac in the liver. Furthermore, a pharmacological intervention that inhibits the H3K4me3 and H3K9ac enrichment resulted in the reversal of statin-induced upregulation of PCSK9. Combining statin and OICR-9429 or resveratrol improved the overall uptake of LDL by hepatocytes. Together, these findings suggest that statin induces the colocalization of H3K4me3 and H3K9ac to transcribe PCSK9 actively and that inhibiting these marks reduces PCSK9 expression and ultimately increases hepatocyte LDL uptake. Our study unveils a previously unknown epigenetic mechanism of PCSK9 regulation that may open new avenues to increase statin efficacy in patients and provide a potential therapeutic solution.
Collapse
Affiliation(s)
- Sushmitha Duddu
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Yash T Katakia
- Department of Biological Sciences, Birla Institute of Technology and Science (BITS), Pilani, India
| | - Rituparna Chakrabarti
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Pooja Sharma
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - Praphulla Chandra Shukla
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India; Department of Human Medicine, Carl von Ossietzky University, Oldenburg, Germany.
| |
Collapse
|
8
|
Behzadi P, St Hilaire C. Metabolites and metabolism in vascular calcification: links between adenosine signaling and the methionine cycle. Am J Physiol Heart Circ Physiol 2024; 327:H1361-H1375. [PMID: 39453431 PMCID: PMC11588312 DOI: 10.1152/ajpheart.00267.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
The global population of individuals with cardiovascular disease is expanding, and a key risk factor for major adverse cardiovascular events is vascular calcification. The pathogenesis of cardiovascular calcification is complex and multifaceted, with external cues driving epigenetic, transcriptional, and metabolic changes that promote vascular calcification. This review provides an overview of some of the lesser understood molecular processes involved in vascular calcification and discusses the links between calcification pathogenesis and aspects of adenosine signaling and the methionine pathway; the latter of which salvages the essential amino acid methionine, but also provides the substrate critical for methylation, a modification that regulates the function and activity of DNA and proteins. We explore the complex and dynamic nature of osteogenic reprogramming underlying intimal atherosclerotic calcification and medial arterial calcification (MAC). Atherosclerotic calcification is more widely studied; however, emerging studies now show that MAC is a significant pathology independent from atherosclerosis. Furthermore, we emphasize metabolite and metabolic-modulating factors that influence vascular calcification pathogenesis. Although the contributions of these mechanisms are more well-define in relation to atherosclerotic intimal calcification, understanding these pathways may provide crucial mechanistic insights into MAC and inform future therapeutic approaches. Herein, we highlight the significance of adenosine and methyltransferase pathways as key regulators of vascular calcification pathogenesis.
Collapse
Affiliation(s)
- Parya Behzadi
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Cynthia St Hilaire
- Division of Cardiology, Department of Medicine, Pittsburgh Heart, Lung, and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
9
|
Wu L, Jiang S, Zhou X, Li W, Ke J, Liu Z, Ren L, Lu Q, Li F, Tang C, Zhu L. Endothelial KDM5B Regulated by Piezo1 Contributes to Disturbed Flow Induced Atherosclerotic Plaque Formation. J Cell Mol Med 2024; 28:e70237. [PMID: 39643939 PMCID: PMC11624123 DOI: 10.1111/jcmm.70237] [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: 07/30/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 12/09/2024] Open
Abstract
Epigenetic modifications play an important role in disturbed flow (d-flow) induced atherosclerotic plaque formation. By analysing a scRNA-seq dataset of the left carotid artery (LCA) under d-flow conditions, we found that Jarid1b (KDM5B) was upregulated primarily in a subcluster of endothelial cells in response to d-flow stimulation. We therefore investigated the mechanism of KDM5B expression and the role of KDM5B in endothelial cell. Intriguingly, activation of Piezo1, a major endothelial mechanosensor, was found to promote KDM5B expression, which was reversed by Piezo1 inhibition in HUVECs. Downstream of Piezo1, ETS1 expression and c-JUN phosphorylation were enhanced by d-flow or Piezo1 activation, leading to an increase in KDM5B expression. Furthermore, knockdown of either KDM5B or Piezo1 was found to prevent d-flow induced H3K4me3 demethylation, which was supported by the pharmacological inhibition of Piezo1 in HUVECs. RNA sequencing on shKdm5b HUVECs implied that KDM5B is associated with endothelial inflammation and atherosclerosis. Using partial carotid ligation surgery on Kdm5bf/f Cdh5cre mice with mAAV-PCSK9D377Y infected, we showed that endothelial KDM5B deficiency reduced atherosclerotic lesions in hypercholesterolemic mice. Our findings indicate that endothelial KDM5B expression induced by d-flow via the Piezo1 pathway promotes atherosclerotic plaque formation, providing targets for the prevention or therapeutic intervention of atherosclerosis.
Collapse
Affiliation(s)
- Lili Wu
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
| | - Shanshan Jiang
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
| | - Xiao Zhou
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
| | - Wei Li
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
| | - Jiaqi Ke
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
| | - Ziting Liu
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
| | - Lijie Ren
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
| | - Qiongyu Lu
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
| | - Fengchan Li
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
| | - Chaojun Tang
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
- Collaborative Innovation Center of Hematology of Jiangsu ProvinceSoochow UniversityJiangsu ProvinceChina
- Suzhou Key Laboratory of Thrombosis and Vascular BiologySuzhouChina
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Li Zhu
- Cyrus Tang Medical InstituteSoochow UniversitySuzhouChina
- Collaborative Innovation Center of Hematology of Jiangsu ProvinceSoochow UniversityJiangsu ProvinceChina
- Suzhou Key Laboratory of Thrombosis and Vascular BiologySuzhouChina
- National Clinical Research Center for Hematologic DiseasesThe First Affiliated Hospital of Soochow UniversitySuzhouChina
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric DiseasesSoochow UniversitySuzhouChina
- Suzhou Ninth Hospital affiliated to Soochow UniversitySoochow UniversitySuzhouChina
| |
Collapse
|
10
|
Al-Awar A, Hussain S. Interplay of Reactive Oxygen Species (ROS) and Epigenetic Remodelling in Cardiovascular Diseases Pathogenesis: A Contemporary Perspective. FRONT BIOSCI-LANDMRK 2024; 29:398. [PMID: 39614429 DOI: 10.31083/j.fbl2911398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/20/2024] [Accepted: 07/24/2024] [Indexed: 12/01/2024]
Abstract
Cardiovascular diseases (CVDs) continue to be the leading cause of mortality worldwide, necessitating the development of novel therapies. Despite therapeutic advancements, the underlying mechanisms remain elusive. Reactive oxygen species (ROS) show detrimental effects at high concentrations but act as essential signalling molecules at physiological levels, playing a critical role in the pathophysiology of CVD. However, the link between pathologically elevated ROS and CVDs pathogenesis remains poorly understood. Recent research has highlighted the remodelling of the epigenetic landscape as a crucial factor in CVD pathologies. Epigenetic changes encompass alterations in DNA methylation, post-translational histone modifications, adenosine triphosphate (ATP)-dependent chromatin modifications, and noncoding RNA transcripts. Unravelling the intricate link between ROS and epigenetic changes in CVD is challenging due to the complexity of epigenetic signals in gene regulation. This review aims to provide insights into the role of ROS in modulating the epigenetic landscape within the cardiovascular system. Understanding these interactions may offer novel therapeutic strategies for managing CVD by targeting ROS-induced epigenetic changes. It has been widely accepted that epigenetic modifications are established during development and remain fixed once the lineage-specific gene expression pattern is achieved. However, emerging evidence has unveiled its remarkable dynamism. Consequently, it is now increasingly recognized that epigenetic modifications may serve as a crucial link between ROS and the underlying mechanisms implicated in CVD.
Collapse
Affiliation(s)
- Amin Al-Awar
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, 41345 Gothenburg, Sweden
| | - Shafaat Hussain
- Department of Molecular and Clinical Medicine, Institute of Medicine, Gothenburg University, 41345 Gothenburg, Sweden
| |
Collapse
|
11
|
Mierke CT. Mechanosensory entities and functionality of endothelial cells. Front Cell Dev Biol 2024; 12:1446452. [PMID: 39507419 PMCID: PMC11538060 DOI: 10.3389/fcell.2024.1446452] [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: 06/09/2024] [Accepted: 10/04/2024] [Indexed: 11/08/2024] Open
Abstract
The endothelial cells of the blood circulation are exposed to hemodynamic forces, such as cyclic strain, hydrostatic forces, and shear stress caused by the blood fluid's frictional force. Endothelial cells perceive mechanical forces via mechanosensors and thus elicit physiological reactions such as alterations in vessel width. The mechanosensors considered comprise ion channels, structures linked to the plasma membrane, cytoskeletal spectrin scaffold, mechanoreceptors, and junctional proteins. This review focuses on endothelial mechanosensors and how they alter the vascular functions of endothelial cells. The current state of knowledge on the dysregulation of endothelial mechanosensitivity in disease is briefly presented. The interplay in mechanical perception between endothelial cells and vascular smooth muscle cells is briefly outlined. Finally, future research avenues are highlighted, which are necessary to overcome existing limitations.
Collapse
|
12
|
Aziz M, Jandeleit-Dahm KA, Khan AW. Interplay between epigenetic mechanisms and transcription factors in atherosclerosis. Atherosclerosis 2024; 395:117615. [PMID: 38917706 DOI: 10.1016/j.atherosclerosis.2024.117615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 05/26/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024]
Abstract
Cardiovascular diseases (CVD), including coronary heart disease and stroke, comprise the number one cause of mortality worldwide. A major contributor to CVD is atherosclerosis, which is a low-grade inflammatory disease of vasculature that involves a pathological build-up of plaque within the arterial walls. Studies have shown that regulation of gene expression via transcription factors and epigenetic mechanisms play a fundamental role in transcriptomic changes linked to the development of atherosclerosis. Chromatin remodeling is a reversible phenomenon and studies have supported the clinical application of chromatin-modifying agents for the prevention and treatment of CVD. In addition, pre-clinical studies have identified multiple transcription factors as potential therapeutic targets in combating atherosclerotic CVD. Although interaction between transcription factors and epigenetic mechanisms facilitate gene regulation, a limited number of studies appreciate this crosstalk in the context of CVD. Here, we reviewed this gene regulatory mechanism underappreciated in atherosclerosis, which will highlight the mechanisms underlying novel therapeutics targeting epigenetic modifiers and transcription factors in atherosclerosis.
Collapse
Affiliation(s)
- Misbah Aziz
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Karin Am Jandeleit-Dahm
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia; German Diabetes Centre, Leibniz Centre for Diabetes Research at the Heinrich Heine University, Dusseldorf, Germany
| | - Abdul Waheed Khan
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia.
| |
Collapse
|
13
|
Sánchez-Ceinos J, Hussain S, Khan AW, Zhang L, Almahmeed W, Pernow J, Cosentino F. Repressive H3K27me3 drives hyperglycemia-induced oxidative and inflammatory transcriptional programs in human endothelium. Cardiovasc Diabetol 2024; 23:122. [PMID: 38580969 PMCID: PMC10998410 DOI: 10.1186/s12933-024-02196-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 03/11/2024] [Indexed: 04/07/2024] Open
Abstract
BACKGROUND Histone modifications play a critical role in chromatin remodelling and regulate gene expression in health and disease. Histone methyltransferases EZH1, EZH2, and demethylases UTX, JMJD3, and UTY catalyse trimethylation of lysine 27 on histone H3 (H3K27me3). This study was designed to investigate whether H3K27me3 triggers hyperglycemia-induced oxidative and inflammatory transcriptional programs in the endothelium. METHODS We studied human aortic endothelial cells exposed to high glucose (HAEC) or isolated from individuals with diabetes (D-HAEC). RT-qPCR, immunoblotting, chromatin immunoprecipitation (ChIP-qPCR), and confocal microscopy were performed to investigate the role of H3K27me3. We determined superoxide anion (O2-) production by ESR spectroscopy, NF-κB binding activity, and monocyte adhesion. Silencing/overexpression and pharmacological inhibition of chromatin modifying enzymes were used to modulate H3K27me3 levels. Furthermore, isometric tension studies and immunohistochemistry were performed in aorta from wild-type and db/db mice. RESULTS Incubation of HAEC to high glucose showed that upregulation of EZH2 coupled to reduced demethylase UTX and JMJD3 was responsible for the increased H3K27me3. ChIP-qPCR revealed that repressive H3K27me3 binding to superoxide dismutase and transcription factor JunD promoters is involved in glucose-induced O2- generation. Indeed, loss of JunD transcriptional inhibition favours NOX4 expression. Furthermore, H3K27me3-driven oxidative stress increased NF-κB p65 activity and downstream inflammatory genes. Interestingly, EZH2 inhibitor GSK126 rescued these endothelial derangements by reducing H3K27me3. We also found that H3K27me3 epigenetic signature alters transcriptional programs in D-HAEC and aortas from db/db mice. CONCLUSIONS EZH2-mediated H3K27me3 represents a key epigenetic driver of hyperglycemia-induced endothelial dysfunction. Targeting EZH2 may attenuate oxidative stress and inflammation and, hence, prevent vascular disease in diabetes.
Collapse
Affiliation(s)
- Julia Sánchez-Ceinos
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Shafaat Hussain
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Abdul Waheed Khan
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Diabetes, Central Clinical School, Monash University, Melbourne, Australia
| | - Liang Zhang
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, UAE
| | - John Pernow
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Francesco Cosentino
- Cardiology Unit, Department of Medicine-Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
14
|
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
|
15
|
Luo X, Shi J, Wang R, Cao L, Gao Y, Wang J, Hong M, Sun X, Zhang Y. Near-Infrared Persistent Luminescence Nanoprobe for Early Detection of Atherosclerotic Plaque. ACS NANO 2024; 18:6500-6512. [PMID: 38348833 DOI: 10.1021/acsnano.3c12136] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Atherosclerosis (AS) is a crucial contributor to various cardiovascular diseases (CVDs), which seriously threaten human life and health. Early and accurate recognition of AS plaques is essential for the prevention and treatment of CVD. Herein, we introduce an AS-targeting nanoprobe based on near-infrared (NIR) persistent luminescence nanoparticles (PLNPs), developing a highly sensitive NIR persistent luminescence (PersL) AS plaque imaging technique and successfully realizing early AS plaque detection. The nanoprobe exhibits good monodispersity and regular spherical morphology and also owns exceptional NIR PersL performance upon repetitive irradiation by biological window light. The surface-conjugated antibody (anti-osteopontin) endowed nanoprobe excellent targeting ability to foam cells within plaques. After intravenously injected nanoprobe into AS model mice, the highly sensitive PersL imaging technique can accurately detect AS plaques prior to ultrasonography (US) and magnetic resonance imaging (MRI). Specifically, the NIR PersL imaging reveals AS plaques at the earliest within 2 weeks, with higher signal-to-background ratio (SBR) up to 5.72. Based on this technique, the nanoprobe has great potential for applications in the prevention and treatment of CVD, the study of AS pathogenesis, and the screening of anti-AS drugs.
Collapse
Affiliation(s)
- Xiaofang Luo
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
- School of Rare Earths, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Science, Ganzhou 341000, People's Republic of China
| | - Junpeng Shi
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
- School of Rare Earths, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Science, Ganzhou 341000, People's Republic of China
| | - Ruoping Wang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| | - Longlong Cao
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| | - Yan Gao
- School of Rare Earths, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Science, Ganzhou 341000, People's Republic of China
| | - Jinyuan Wang
- School of Rare Earths, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Ganjiang Innovation Academy, Chinese Academy of Science, Ganzhou 341000, People's Republic of China
| | - Maochun Hong
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| | - Xia Sun
- Fujian Science and Technology Innovation Laboratory for Optoelectronic Information of China, Fuzhou 350108, People's Republic of China
| | - Yun Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, People's Republic of China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, People's Republic of China
| |
Collapse
|
16
|
Raghubeer S. The influence of epigenetics and inflammation on cardiometabolic risks. Semin Cell Dev Biol 2024; 154:175-184. [PMID: 36804178 DOI: 10.1016/j.semcdb.2023.02.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023]
Abstract
Cardiometabolic diseases include metabolic syndrome, obesity, type 2 diabetes mellitus, and hypertension. Epigenetic modifications participate in cardiometabolic diseases through several pathways, including inflammation, vascular dysfunction, and insulin resistance. Epigenetic modifications, which encompass alterations to gene expression without mutating the DNA sequence, have gained much attention in recent years, since they have been correlated with cardiometabolic diseases and may be targeted for therapeutic interventions. Epigenetic modifications are greatly influenced by environmental factors, such as diet, physical activity, cigarette smoking, and pollution. Some modifications are heritable, indicating that the biological expression of epigenetic alterations may be observed across generations. Moreover, many patients with cardiometabolic diseases present with chronic inflammation, which can be influenced by environmental and genetic factors. The inflammatory environment worsens the prognosis of cardiometabolic diseases and further induces epigenetic modifications, predisposing patients to the development of other metabolism-associated diseases and complications. A deeper understanding of inflammatory processes and epigenetic modifications in cardiometabolic diseases is necessary to improve our diagnostic capabilities, personalized medicine approaches, and the development of targeted therapeutic interventions. Further understanding may also assist in predicting disease outcomes, especially in children and young adults. This review describes epigenetic modifications and inflammatory processes underlying cardiometabolic diseases, and further discusses advances in the research field with a focus on specific points for interventional therapy.
Collapse
Affiliation(s)
- Shanel Raghubeer
- SAMRC/CPUT/Cardiometabolic Health Research Unit, Department of Biomedical Sciences, Faculty of Health & Wellness Sciences, Cape Peninsula University of Technology, South Africa.
| |
Collapse
|
17
|
Jazieh C, Arabi TZ, Asim Z, Sabbah BN, Alsaud AW, Alkattan K, Yaqinuddin A. Unraveling the epigenetic fabric of type 2 diabetes mellitus: pathogenic mechanisms and therapeutic implications. Front Endocrinol (Lausanne) 2024; 15:1295967. [PMID: 38323108 PMCID: PMC10845351 DOI: 10.3389/fendo.2024.1295967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a rapidly escalating global health concern, with its prevalence projected to increase significantly in the near future. This review delves into the intricate role of epigenetic modifications - including DNA methylation, histone acetylation, and micro-ribonucleic acid (miRNA) expression - in the pathogenesis and progression of T2DM. We critically examine how these epigenetic changes contribute to the onset and exacerbation of T2DM by influencing key pathogenic processes such as obesity, insulin resistance, β-cell dysfunction, cellular senescence, and mitochondrial dysfunction. Furthermore, we explore the involvement of epigenetic dysregulation in T2DM-associated complications, including diabetic retinopathy, atherosclerosis, neuropathy, and cardiomyopathy. This review highlights recent studies that underscore the diagnostic and therapeutic potential of targeting epigenetic modifications in T2DM. We also provide an overview of the impact of lifestyle factors such as exercise and diet on the epigenetic landscape of T2DM, underscoring their relevance in disease management. Our synthesis of the current literature aims to illuminate the complex epigenetic underpinnings of T2DM, offering insights into novel preventative and therapeutic strategies that could revolutionize its management.
Collapse
|
18
|
Kumar K, Anjali S, Sharma S. Effect of lead exposure on histone modifications: A review. J Biochem Mol Toxicol 2024; 38:e23547. [PMID: 37867311 DOI: 10.1002/jbt.23547] [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: 06/14/2023] [Revised: 08/21/2023] [Accepted: 09/26/2023] [Indexed: 10/24/2023]
Abstract
Lead at any levels can result in detrimental health effects affecting various organ systems. These systematic manifestations under Pb exposure and the underlying probable pathophysiological mechanisms have not been elucidated completely. With advancements in molecular research under Pb exposure, epigenetics is one of the emerging field that has opened many possibilities for appreciating the role of Pb exposure in modulating gene expression profiles. In terms of epigenetic alterations reported in Pb toxicity, DNA methylation, and microRNA alterations are extensively explored in both experimental and epidemiological studies, however, the understanding of histone modifications under Pb exposure is still in its infant stage limited to experimental models. In this review, we aim to present a synoptic view of histone modifications explored in relation to Pb exposure attempting to bring out this potential lacunae in research. The scarcity of studies associating histone modifications with Pb toxicity, and the paucity of their validation in human cohort further emphasizes the strong research potential of this field. We summarize the review by presenting our hypotheses regarding the involvement of these histone modification in various diseases modalities associated with Pb toxicity.
Collapse
Affiliation(s)
- Kanishka Kumar
- Department of Biochemistry, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| | - Sudha Anjali
- Department of Biochemistry, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| | - Shailja Sharma
- Department of Biochemistry, AIIMS Jodhpur, Jodhpur, Rajasthan, India
| |
Collapse
|
19
|
Gladwell LR, Ahiarah C, Rasheed S, Rahman SM, Choudhury M. Traditional Therapeutics and Potential Epidrugs for CVD: Why Not Both? Life (Basel) 2023; 14:23. [PMID: 38255639 PMCID: PMC10820772 DOI: 10.3390/life14010023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/18/2023] [Indexed: 01/24/2024] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. In addition to the high mortality rate, people suffering from CVD often endure difficulties with physical activities and productivity that significantly affect their quality of life. The high prevalence of debilitating risk factors such as obesity, type 2 diabetes mellitus, smoking, hypertension, and hyperlipidemia only predicts a bleak future. Current traditional CVD interventions offer temporary respite; however, they compound the severe economic strain of health-related expenditures. Furthermore, these therapeutics can be prescribed indefinitely. Recent advances in the field of epigenetics have generated new treatment options by confronting CVD at an epigenetic level. This involves modulating gene expression by altering the organization of our genome rather than altering the DNA sequence itself. Epigenetic changes are heritable, reversible, and influenced by environmental factors such as medications. As CVD is physiologically and pathologically diverse in nature, epigenetic interventions can offer a ray of hope to replace or be combined with traditional therapeutics to provide the prospect of addressing more than just the symptoms of CVD. This review discusses various risk factors contributing to CVD, perspectives of current traditional medications in practice, and a focus on potential epigenetic therapeutics to be used as alternatives.
Collapse
Affiliation(s)
- Lauren Rae Gladwell
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Chidinma Ahiarah
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Shireen Rasheed
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| | - Shaikh Mizanoor Rahman
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mouz, Nizwa 616, Oman
| | - Mahua Choudhury
- Department of Pharmaceutical Sciences, Texas A&M Irma Lerma Rangel College of Pharmacy, 1114 TAMU, College Station, TX 77843, USA
| |
Collapse
|
20
|
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: 15] [Impact Index Per Article: 7.5] [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
|
21
|
Bergonzini M, Loreni F, Lio A, Russo M, Saitto G, Cammardella A, Irace F, Tramontin C, Chello M, Lusini M, Nenna A, Ferrisi C, Ranocchi F, Musumeci F. Panoramic on Epigenetics in Coronary Artery Disease and the Approach of Personalized Medicine. Biomedicines 2023; 11:2864. [PMID: 37893238 PMCID: PMC10604795 DOI: 10.3390/biomedicines11102864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Epigenetic modifications play a fundamental role in the progression of coronary artery disease (CAD). This panoramic review aims to provide an overview of the current understanding of the epigenetic mechanisms involved in CAD pathogenesis and highlights the potential implications for personalized medicine approaches. Epigenetics is the study of heritable changes that do not influence alterations in the DNA sequence of the genome. It has been shown that epigenetic processes, including DNA/histone methylation, acetylation, and phosphorylation, play an important role. Additionally, miRNAs, lncRNAs, and circRNAs are also involved in epigenetics, regulating gene expression patterns in response to various environmental factors and lifestyle choices. In the context of CAD, epigenetic alterations contribute to the dysregulation of genes involved in inflammation, oxidative stress, lipid metabolism, and vascular function. These epigenetic changes can occur during early developmental stages and persist throughout life, predisposing individuals to an increased risk of CAD. Furthermore, in recent years, the concept of personalized medicine has gained significant attention. Personalized medicine aims to tailor medical interventions based on an individual's unique genetic, epigenetic, environmental, and lifestyle factors. In the context of CAD, understanding the interplay between genetic variants and epigenetic modifications holds promise for the development of more precise diagnostic tools, risk stratification models, and targeted therapies. This review summarizes the current knowledge of epigenetic mechanisms in CAD and discusses the fundamental principles of personalized medicine.
Collapse
Affiliation(s)
- Marcello Bergonzini
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Francesco Loreni
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Antonio Lio
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Marco Russo
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Guglielmo Saitto
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Antonio Cammardella
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Francesco Irace
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Corrado Tramontin
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Massimo Chello
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Mario Lusini
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Antonio Nenna
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Chiara Ferrisi
- Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Federico Ranocchi
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| | - Francesco Musumeci
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, 00152 Rome, Italy
| |
Collapse
|
22
|
Aronova A, Tosato F, Naser N, Asare Y. Innate Immune Pathways in Atherosclerosis-From Signaling to Long-Term Epigenetic Reprogramming. Cells 2023; 12:2359. [PMID: 37830572 PMCID: PMC10571887 DOI: 10.3390/cells12192359] [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: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Innate immune pathways play a crucial role in the development of atherosclerosis, from sensing initial danger signals to the long-term reprogramming of immune cells. Despite the success of lipid-lowering therapy, anti-hypertensive medications, and other measures in reducing complications associated with atherosclerosis, cardiovascular disease (CVD) remains the leading cause of death worldwide. Consequently, there is an urgent need to devise novel preventive and therapeutic strategies to alleviate the global burden of CVD. Extensive experimental research and epidemiological studies have demonstrated the dominant role of innate immune mechanisms in the progression of atherosclerosis. Recently, landmark trials including CANTOS, COLCOT, and LoDoCo2 have provided solid evidence demonstrating that targeting innate immune pathways can effectively reduce the risk of CVD. These groundbreaking trials mark a significant paradigm shift in the field and open new avenues for atheroprotective treatments. It is therefore crucial to comprehend the intricate interplay between innate immune pathways and atherosclerosis for the development of targeted therapeutic interventions. Additionally, unraveling the mechanisms underlying long-term reprogramming may offer novel strategies to reverse the pro-inflammatory phenotype of immune cells and restore immune homeostasis in atherosclerosis. In this review, we present an overview of the innate immune pathways implicated in atherosclerosis, with a specific focus on the signaling pathways driving chronic inflammation in atherosclerosis and the long-term reprogramming of immune cells within atherosclerotic plaque. Elucidating the molecular mechanisms governing these processes presents exciting opportunities for the development of a new class of immunotherapeutic approaches aimed at reducing inflammation and promoting plaque stability. By addressing these aspects, we can potentially revolutionize the management of atherosclerosis and its associated cardiovascular complications.
Collapse
Affiliation(s)
| | | | | | - Yaw Asare
- Institute for Stroke and Dementia Research (ISD), University Hospital, Ludwig-Maximilian-University (LMU), 80539 Munich, Germany
| |
Collapse
|
23
|
Forman DE, Kuchel GA, Newman JC, Kirkland JL, Volpi E, Taffet GE, Barzilai N, Pandey A, Kitzman DW, Libby P, Ferrucci L. Impact of Geroscience on Therapeutic Strategies for Older Adults With Cardiovascular Disease: JACC Scientific Statement. J Am Coll Cardiol 2023; 82:631-647. [PMID: 37389519 PMCID: PMC10414756 DOI: 10.1016/j.jacc.2023.05.038] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/09/2023] [Accepted: 05/31/2023] [Indexed: 07/01/2023]
Abstract
Geroscience posits that cardiovascular disease (CVD) and other chronic diseases result from progressive erosion of the effectiveness of homeostatic mechanisms that oppose age-related accumulation of molecular damage. This hypothetical common root to chronic diseases explains why patients with CVD are often affected by multimorbidity and frailty and why older age negatively affects CVD prognosis and treatment response. Gerotherapeutics enhance resilience mechanisms that counter age-related molecular damage to prevent chronic diseases, frailty, and disability, thereby extending healthspan. Here, we describe the main resilience mechanisms of mammalian aging, with a focus on how they can affect CVD pathophysiology. We next present novel gerotherapeutic approaches, some of which are already used in management of CVD, and explore their potential to transform care and management of CVD. The geroscience paradigm is gaining traction broadly in medical specialties, with potential to mitigate premature aging, reduce health care disparities, and improve population healthspan.
Collapse
Affiliation(s)
- Daniel E Forman
- Department of Medicine (Geriatrics and Cardiology) University of Pittsburgh, Pittsburgh, Pennsylvania, USA; GRECC, VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, USA.
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut School of Medicine, UConn Health, Farmington, Connecticut, USA
| | - John C Newman
- Buck Institute for Research on Aging, Novato California, USA; Division of Geriatrics, University of California San Francisco, San Francisco, California, USA
| | - James L Kirkland
- Division of General Internal Medicine, Department of Medicine and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Elena Volpi
- Sealy Center on Aging, University of Texas Medical Branch, Galveston, Texas, USA
| | - George E Taffet
- Department of Medicine (Geriatrics and Cardiovascular Sciences), Baylor College of Medicine, Houston, Texas, USA
| | - Nir Barzilai
- Einstein Institute for Aging Research, Bronx, New York, USA; Einstein-NSC and Glenn Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Ambarish Pandey
- Division of Cardiology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Dalane W Kitzman
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Peter Libby
- Cardiovascular Medicine and Geriatrics, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | | |
Collapse
|
24
|
Farsetti A, Illi B, Gaetano C. How epigenetics impacts on human diseases. Eur J Intern Med 2023; 114:15-22. [PMID: 37277249 DOI: 10.1016/j.ejim.2023.05.036] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/07/2023]
Abstract
Epigenetics is a rapidly growing field of biology that studies the changes in gene expression that are not due to alterations in the DNA sequence but rather the chemical modifications of DNA and its associated proteins. Epigenetic mechanisms can profoundly influence gene expression, cell differentiation, tissue development, and disease susceptibility. Understanding epigenetic changes is essential to elucidate the mechanisms underlying the increasingly recognized role of environmental and lifestyle factors in health and disease and the intergenerational transmission of phenotypes. Recent studies suggest epigenetics may be critical in various diseases, from cardiovascular disease and cancer to neurodevelopmental and neurodegenerative disorders. Epigenetic modifications are potentially reversible and could provide new therapeutic avenues for treating these diseases using epigenetic modulators. Moreover, epigenetics provide insight into disease pathogenesis and biomarkers for disease diagnosis and risk stratification. Nevertheless, epigenetic interventions have the potential for unintended consequences and may potentially lead to increased risks of unexpected outcomes, such as adverse drug reactions, developmental abnormalities, and cancer. Therefore, rigorous studies are essential to minimize the risks associated with epigenetic therapies and to develop safe and effective interventions for improving human health. This article provides a synthetic and historical view of the origin of epigenetics and some of the most relevant achievements.
Collapse
Affiliation(s)
- Antonella Farsetti
- Istituto di analisi dei sistemi ed informatica "Antonio Ruberti" (IASI), Consiglio Nazionale delle Ricerche (CNR), Via dei Taurini, 19 - 00185 Roma, Italy
| | - Barbara Illi
- Istituto di biologia e Patologia Molecolari, (IBPM), Consiglio Nazionale delle Ricerche (CNR), P.le Aldo Moro 5, 00185, Roma, Italy
| | - Carlo Gaetano
- Laboratorio di Epigenetica, Istituti Cinici Scientifici Maugeri IRCCS, Via Maugeri 4, 27100, Pavia, Italy.
| |
Collapse
|
25
|
Sopic M, Robinson EL, Emanueli C, Srivastava P, Angione C, Gaetano C, Condorelli G, Martelli F, Pedrazzini T, Devaux Y. Integration of epigenetic regulatory mechanisms in heart failure. Basic Res Cardiol 2023; 118:16. [PMID: 37140699 PMCID: PMC10158703 DOI: 10.1007/s00395-023-00986-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 03/27/2023] [Accepted: 04/10/2023] [Indexed: 05/05/2023]
Abstract
The number of "omics" approaches is continuously growing. Among others, epigenetics has appeared as an attractive area of investigation by the cardiovascular research community, notably considering its association with disease development. Complex diseases such as cardiovascular diseases have to be tackled using methods integrating different omics levels, so called "multi-omics" approaches. These approaches combine and co-analyze different levels of disease regulation. In this review, we present and discuss the role of epigenetic mechanisms in regulating gene expression and provide an integrated view of how these mechanisms are interlinked and regulate the development of cardiac disease, with a particular attention to heart failure. We focus on DNA, histone, and RNA modifications, and discuss the current methods and tools used for data integration and analysis. Enhancing the knowledge of these regulatory mechanisms may lead to novel therapeutic approaches and biomarkers for precision healthcare and improved clinical outcomes.
Collapse
Affiliation(s)
- Miron Sopic
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Emma L Robinson
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Costanza Emanueli
- National Heart & Lung Institute, Imperial College London, London, UK
| | | | - Claudio Angione
- School of Computing, Engineering & Digital Technologies, Teesside University, Tees Valley, Middlesbrough, TS1 3BA, UK
- Centre for Digital Innovation, Teesside University, Campus Heart, Tees Valley, Middlesbrough, TS1 3BX, UK
- National Horizons Centre, Darlington, DL1 1HG, UK
| | - Carlo Gaetano
- Laboratorio di Epigenetica, Istituti Clinici Scientifici Maugeri IRCCS, Via Maugeri 10, 27100, Pavia, Italy
| | - Gianluigi Condorelli
- IRCCS-Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, MI, Italy
- Institute of Genetic and Biomedical Research, National Research Council of Italy, Arnold-Heller-Str.3, 24105, Milan, Italy
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, Via Morandi 30, San Donato Milanese, 20097, Milan, Italy
| | - Thierry Pedrazzini
- Experimental Cardiology Unit, Division of Cardiology, Department of Cardiovascular Medicine, University of Lausanne Medical School, 1011, Lausanne, Switzerland
| | - Yvan Devaux
- Cardiovascular Research Unit, Department of Population Health, Luxembourg Institute of Health, L-1445, Strassen, Luxembourg.
| |
Collapse
|
26
|
Mannar V, Boro H, Patel D, Agstam S, Dalvi M, Bundela V. Epigenetics of the Pathogenesis and Complications of Type 2 Diabetes Mellitus. TOUCHREVIEWS IN ENDOCRINOLOGY 2023; 19:46-53. [PMID: 37313245 PMCID: PMC10258626 DOI: 10.17925/ee.2023.19.1.46] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 02/23/2023] [Indexed: 06/15/2023]
Abstract
Epigenetics of type 2 diabetes mellitus (T2DM) has widened our knowledge of various aspects of the disease. The aim of this review is to summarize the important epigenetic changes implicated in the disease risks, pathogenesis, complications and the evolution of therapeutics in our current understanding of T2DM. Studies published in the past 15 years, from 2007 to 2022, from three primary platforms namely PubMed, Google Scholar and Science Direct were included. Studies were searched using the primary term 'type 2 diabetes and epigenetics' with additional terms such as 'risks', 'pathogenesis', 'complications of diabetes' and 'therapeutics'. Epigenetics plays an important role in the transmission of T2DM from one generation to another. Epigenetic changes are also implicated in the two basic pathogenic components of T2DM, namely insulin resistance and impaired insulin secretion. Hyperglycaemia-i nduced permanent epigenetic modifications of the expression of DNA are responsible for the phenomenon of metabolic memory. Epigenetics influences the development of micro-and macrovascular complications of T2DM. They can also be used as biomarkers in the prediction of these complications. Epigenetics has expanded our understanding of the action of existing drugs such as metformin, and has led to the development of newer targets to prevent vascular complications. Epigenetic changes are involved in almost all aspects of T2DM, from risks, pathogenesis and complications, to the development of newer therapeutic targets.
Collapse
Affiliation(s)
- Velmurugan Mannar
- Department of Medicine, Aarupadai Veedu Medical College, Puducherry, India
| | - Hiya Boro
- Department of Endocrinology and Metabolism, Aadhar Health Institute, Hisar, India
| | - Deepika Patel
- Department of Endocrinology, Mediheal Hospital, Nairobi, Kenya
| | - Sourabh Agstam
- Department of Cardiology, VMMC and Safdarjung Hospital, New Delhi, India
| | - Mazhar Dalvi
- Department of Endocrinology, Mediclinic Al Noor Hospital, Abu Dhabi, United Arab Emirates
| | - Vikash Bundela
- Department of Gastroenterology, Aadhar Health Institute, Hisar, India
| |
Collapse
|
27
|
Sharma SB, Melvin WJ, Audu CO, Bame M, Rhoads N, Wu W, Kanthi Y, Knight JS, Adili R, Holinstat MA, Wakefield TW, Henke PK, Moore BB, Gallagher KA, Obi AT. The histone methyltransferase MLL1/KMT2A in monocytes drives coronavirus-associated coagulopathy and inflammation. Blood 2023; 141:725-742. [PMID: 36493338 PMCID: PMC9743412 DOI: 10.1182/blood.2022015917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 12/13/2022] Open
Abstract
Coronavirus-associated coagulopathy (CAC) is a morbid and lethal sequela of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. CAC results from a perturbed balance between coagulation and fibrinolysis and occurs in conjunction with exaggerated activation of monocytes/macrophages (MO/Mφs), and the mechanisms that collectively govern this phenotype seen in CAC remain unclear. Here, using experimental models that use the murine betacoronavirus MHVA59, a well-established model of SARS-CoV-2 infection, we identify that the histone methyltransferase mixed lineage leukemia 1 (MLL1/KMT2A) is an important regulator of MO/Mφ expression of procoagulant and profibrinolytic factors such as tissue factor (F3; TF), urokinase (PLAU), and urokinase receptor (PLAUR) (herein, "coagulopathy-related factors") in noninfected and infected cells. We show that MLL1 concurrently promotes the expression of the proinflammatory cytokines while suppressing the expression of interferon alfa (IFN-α), a well-known inducer of TF and PLAUR. Using in vitro models, we identify MLL1-dependent NF-κB/RelA-mediated transcription of these coagulation-related factors and identify a context-dependent, MLL1-independent role for RelA in the expression of these factors in vivo. As functional correlates for these findings, we demonstrate that the inflammatory, procoagulant, and profibrinolytic phenotypes seen in vivo after coronavirus infection were MLL1-dependent despite blunted Ifna induction in MO/Mφs. Finally, in an analysis of SARS-CoV-2 positive human samples, we identify differential upregulation of MLL1 and coagulopathy-related factor expression and activity in CD14+ MO/Mφs relative to noninfected and healthy controls. We also observed elevated plasma PLAU and TF activity in COVID-positive samples. Collectively, these findings highlight an important role for MO/Mφ MLL1 in promoting CAC and inflammation.
Collapse
Affiliation(s)
- Sriganesh B. Sharma
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - William J. Melvin
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Christopher O. Audu
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Monica Bame
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| | - Nicole Rhoads
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Weisheng Wu
- Bioinformatics Core, Biomedical Research Core Facilities, University of Michigan, Ann Arbor, MI
| | - Yogendra Kanthi
- Laboratory of Vascular Thrombosis & Inflammation, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Jason S. Knight
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Reheman Adili
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Michael A. Holinstat
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Department of Pharmacology, University of Michigan, Ann Arbor, MI
| | - Thomas W. Wakefield
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Peter K. Henke
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| | - Katherine A. Gallagher
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI
| | - Andrea T. Obi
- Section of General Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
- Section of Vascular Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI
| |
Collapse
|
28
|
Aziz S, Yalan L, Raza MA, Lemin J, Akram HMB, Zhao W. GSK126 an inhibitor of epigenetic regulator EZH2 suppresses cardiac fibrosis by regulating the EZH2-PAX6-CXCL10 pathway. Biochem Cell Biol 2023; 101:87-100. [PMID: 36469862 DOI: 10.1139/bcb-2022-0224] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Myocardial fibrosis is a common pathological companion of various cardiovascular diseases. To date, the role of enhancer of zeste homolog 2 (EZH2) in cancer has been well demonstrated including in renal carcinoma and its inhibitors have entered the stage of phase I/II clinical trials. However, the precise mechanism of EZH2 in cardiac diseases is largely unclear. In the current study, we first found that EZH2 expression was increased in Ang-II-treated cardiac fibroblasts (CFs) and mouse heart homogenates following isoproterenol (ISO) administration for 21 days, respectively. Ang-II induces CFs activation and increased collagen-I, collagen-III, α-SMA, EZH2, and trimethylates lysine 27 on histone 3 (H3K27me3) expressions can be reversed by EZH2 inhibitor (GSK126) and EZH2 siRNA. The ISO-induced cardiac hypertrophy, and fibrosis in vivo which were also related to the upregulation of EZH2 and its downstream target, H3K27me3, could be recovered by GSK126. Furthermore, the upregulation of EZH2 induces the decrease of paired box 6 (PAX6) and C-X-C motif ligand 10 (CXCL10) "which" were also reversed by GSK126 treatment. In summary, the present evidence strongly suggests that GSK126 could be a therapeutic intervention, blunting the development and progression of myocardial fibrosis in an EZH2-PAX6-CXCL10-dependent manner.
Collapse
Affiliation(s)
- Shireen Aziz
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Li Yalan
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Muhammad Ahmer Raza
- Department of Clinical Pharmacy, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiao Lemin
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Hafiz Muhamamd Bilal Akram
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Wen Zhao
- State Key Laboratory of Esophageal Cancer Prevention and Treatment; Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China; School of Pharmaceutical Sciences and Institute of Drug Discovery & Development, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| |
Collapse
|
29
|
Ibarrola J, Kim SK, Lu Q, DuPont JJ, Creech A, Sun Z, Hill MA, Jaffe JD, Jaffe IZ. Smooth muscle mineralocorticoid receptor as an epigenetic regulator of vascular ageing. Cardiovasc Res 2023; 118:3386-3400. [PMID: 35020830 PMCID: PMC10060709 DOI: 10.1093/cvr/cvac007] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 01/07/2022] [Indexed: 01/25/2023] Open
Abstract
AIMS Vascular stiffness increases with age and independently predicts cardiovascular disease risk. Epigenetic changes, including histone modifications, accumulate with age but the global pattern has not been elucidated nor are the regulators known. Smooth muscle cell-mineralocorticoid receptor (SMC-MR) contributes to vascular stiffness in ageing mice. Thus, we investigated the regulatory role of SMC-MR in vascular epigenetics and stiffness. METHODS AND RESULTS Mass spectrometry-based proteomic profiling of all histone modifications completely distinguished 3 from 12-month-old mouse aortas. Histone-H3 lysine-27 (H3K27) methylation (me) significantly decreased in ageing vessels and this was attenuated in SMC-MR-KO littermates. Immunoblotting revealed less H3K27-specific methyltransferase EZH2 with age in MR-intact but not SMC-MR-KO vessels. These ageing changes were examined in primary human aortic (HA)SMC from adult vs. aged donors. MR, H3K27 acetylation (ac), and stiffness gene (connective tissue growth factor, integrin-α5) expression significantly increased, while H3K27me and EZH2 decreased, with age. MR inhibition reversed these ageing changes in HASMC and the decline in stiffness genes was prevented by EZH2 blockade. Atomic force microscopy revealed that MR antagonism decreased intrinsic stiffness and the probability of fibronectin adhesion of aged HASMC. Conversely, ageing induction in young HASMC with H2O2; increased MR, decreased EZH2, enriched H3K27ac and MR at stiffness gene promoters by chromatin immunoprecipitation, and increased stiffness gene expression. In 12-month-old mice, MR antagonism increased aortic EZH2 and H3K27 methylation, increased EZH2 recruitment and decreased H3K27ac at stiffness genes promoters, and prevented ageing-induced vascular stiffness and fibrosis. Finally, in human aortic tissue, age positively correlated with MR and stiffness gene expression and negatively correlated with H3K27me3 while MR and EZH2 are negatively correlated. CONCLUSION These data support a novel vascular ageing model with rising MR in human SMC suppressing EZH2 expression thereby decreasing H3K27me, promoting MR recruitment and H3K27ac at stiffness gene promoters to induce vascular stiffness and suggests new targets for ameliorating ageing-associated vascular disease.
Collapse
Affiliation(s)
- Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Box 80, Boston, MA 02111, USA
| | - Seung Kyum Kim
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Box 80, Boston, MA 02111, USA
- Department of Sports Science, Seoul National University of Science and Technology, 232 Gongneung-ro, Nowon-gu, 01811 Republic of Korea, Seoul, South Korea
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Box 80, Boston, MA 02111, USA
| | - Jennifer J DuPont
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Box 80, Boston, MA 02111, USA
| | - Amanda Creech
- Broad Institute, Proteomics Platform, Cambridge, MA 02142, USA
| | - Zhe Sun
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65203, USA
| | - Michael A Hill
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65203, USA
| | - Jacob D Jaffe
- Broad Institute, Proteomics Platform, Cambridge, MA 02142, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, Box 80, Boston, MA 02111, USA
| |
Collapse
|
30
|
Komal S, Han SN, Cui LG, Zhai MM, Zhou YJ, Wang P, Shakeel M, Zhang LR. Epigenetic Regulation of Macrophage Polarization in Cardiovascular Diseases. Pharmaceuticals (Basel) 2023; 16:141. [PMID: 37259293 PMCID: PMC9963081 DOI: 10.3390/ph16020141] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 08/17/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of hospitalization and death worldwide, especially in developing countries. The increased prevalence rate and mortality due to CVDs, despite the development of several approaches for prevention and treatment, are alarming trends in global health. Chronic inflammation and macrophage infiltration are key regulators of the initiation and progression of CVDs. Recent data suggest that epigenetic modifications, such as DNA methylation, posttranslational histone modifications, and RNA modifications, regulate cell development, DNA damage repair, apoptosis, immunity, calcium signaling, and aging in cardiomyocytes; and are involved in macrophage polarization and contribute significantly to cardiac disease development. Cardiac macrophages not only trigger damaging inflammatory responses during atherosclerotic plaque formation, myocardial injury, and heart failure but are also involved in tissue repair, remodeling, and regeneration. In this review, we summarize the key epigenetic modifications that influence macrophage polarization and contribute to the pathophysiology of CVDs, and highlight their potential for the development of advanced epigenetic therapies.
Collapse
Affiliation(s)
- Sumra Komal
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Sheng-Na Han
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Liu-Gen Cui
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Miao-Miao Zhai
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yue-Jiao Zhou
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Pei Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Muhammad Shakeel
- Jamil-ur-Rahman Center for Genome Research, Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Li-Rong Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
31
|
Wang E, Wang H, Chakrabarti S. Endothelial-to-mesenchymal transition: An underappreciated mediator of diabetic complications. Front Endocrinol (Lausanne) 2023; 14:1050540. [PMID: 36777351 PMCID: PMC9911675 DOI: 10.3389/fendo.2023.1050540] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Diabetes and its complications represent a great burden on the global healthcare system. Diabetic complications are fundamentally diseases of the vasculature, with endothelial cells being the centerpiece of early hyperglycemia-induced changes. Endothelial-to-mesenchymal transition is a tightly regulated process that results in endothelial cells losing endothelial characteristics and developing mesenchymal traits. Although endothelial-to-mesenchymal transition has been found to occur within most of the major complications of diabetes, it has not been a major focus of study or a common target in the treatment or prevention of diabetic complications. In this review we summarize the importance of endothelial-to-mesenchymal transition in each major diabetic complication, examine specific mechanisms at play, and highlight potential mechanisms to prevent endothelial-to-mesenchymal transition in each of the major chronic complications of diabetes.
Collapse
|
32
|
Manea SA, Vlad ML, Lazar AG, Muresian H, Simionescu M, Manea A. Pharmacological Inhibition of Lysine-Specific Demethylase 1A Reduces Atherosclerotic Lesion Formation in Apolipoprotein E-Deficient Mice by a Mechanism Involving Decreased Oxidative Stress and Inflammation; Potential Implications in Human Atherosclerosis. Antioxidants (Basel) 2022; 11:antiox11122382. [PMID: 36552592 PMCID: PMC9774905 DOI: 10.3390/antiox11122382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Dysregulated epigenetic mechanisms promote transcriptomic and phenotypic alterations in cardiovascular diseases. The role of histone methylation-related pathways in atherosclerosis is largely unknown. We hypothesize that lysine-specific demethylase 1A (LSD1/KDM1A) regulates key molecular effectors and pathways linked to atherosclerotic plaque formation. Human non-atherosclerotic and atherosclerotic tissue specimens, ApoE-/- mice, and in vitro polarized macrophages (Mac) were examined. Male ApoE-/- mice fed a normal/atherogenic diet were randomized to receive GSK2879552, a highly specific LSD1 inhibitor, or its vehicle, for 4 weeks. The mRNA and protein expression levels of LSD1/KDM1A were significantly elevated in atherosclerotic human carotid arteries, atherosclerotic aortas of ApoE-/- mice, and M1-Mac. Treatment of ApoE-/- mice with GSK2879552 significantly reduced the extent of atherosclerotic lesions and the aortic expression of NADPH oxidase subunits (Nox1/2/4, p22phox) and 4-hydroxynonenal-protein adducts. Concomitantly, the markers of immune cell infiltration and vascular inflammation were significantly decreased. LSD1 blockade down-regulated the expression of genes associated with Mac pro-inflammatory phenotype. Nox subunit transcript levels were significantly elevated in HEK293 reporter cells overexpressing LSD1. In experimental atherosclerosis, LSD1 mediates the up-regulation of molecular effectors connected to oxidative stress and inflammation. Together, these data indicate that LSD1-pharmacological interventions are novel targets for supportive therapeutic strategies in atherosclerosis.
Collapse
Affiliation(s)
- Simona-Adriana Manea
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania
| | - Mihaela-Loredana Vlad
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania
| | - Alexandra-Gela Lazar
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania
| | - Horia Muresian
- Cardiovascular Surgery Department, University Hospital Bucharest, 050098 Bucharest, Romania
| | - Maya Simionescu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania
| | - Adrian Manea
- Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania
- Correspondence:
| |
Collapse
|
33
|
Chen MY, Zhang ZH, Ke JF, Li TT, Li MF, Lu JX, Li LX. Chaetocin attenuates atherosclerosis progression and inhibits vascular smooth muscle cell phenotype switching. J Cardiovasc Transl Res 2022; 15:1270-1282. [PMID: 35428928 DOI: 10.1007/s12265-022-10258-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 04/06/2022] [Indexed: 12/16/2022]
Abstract
We aimed to explore the effect of chaetocin on atherosclerosis and its possible mechanism. In vitro, we observed that chaetocin treatment significantly inhibited the proliferation of VSMCs in concentration- and time-dependent manner. We also found that chaetocin suppressed the migration of VSMCs. Moreover, chaetocin treatment induced a contractile phenotype in VSMCs by increasing α-SMA and SM22α expression. In addition, chaetocin treatment attenuated the accumulation of H3K9me3 on VSMCs contractile gene promoters, which promoted the expression of α-SMA and SM22α. In vivo, chaetocin treatment decreased the H3K9me3 expression, diminished atherosclerotic plaque formation, and increased plaque stability by decreasing necrotic core area and lipid accumulation and increasing collagen content and contractile VSMC phenotype. We demonstrated a new function of chaetocin in inhibiting atherosclerosis progression and increasing plaque stability partly by inhibiting pathological phenotypic switching of VSMCs. These newly identified roles of chaetocin might provide a novel therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Ming-Yun Chen
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
- Department of Endocrinology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, Fujian, People's Republic of China
| | - Zhi-Hui Zhang
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
| | - Jiang-Feng Ke
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
| | - Ting-Ting Li
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
| | - Mei-Fang Li
- Department of Emergency, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Jun-Xi Lu
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China
| | - Lian-Xi Li
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 200233, Shanghai, People's Republic of China.
| |
Collapse
|
34
|
Protective Effect of Galangin Methylation Modification Based on Cell Imaging on Inflammatory Lung Injury and Its Molecular Mechanism. CONTRAST MEDIA & MOLECULAR IMAGING 2022; 2022:7511345. [PMID: 36072628 PMCID: PMC9398807 DOI: 10.1155/2022/7511345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/21/2022]
Abstract
Background Recently, inflammation has become a major threat to human health. Studies have confirmed that some Chinese traditional medicine ingredients may effectively interfere with the expression of inflammatory mediators through epigenetic modification, showing a great potential of the application. Objective To investigate the role of the PPAR/DNMT3A pathway in the reversal of galangin-mediated inflammatory lung injury, promote the development of new anti-inflammatory drugs, reduce the side effects of chemical synthetic drugs on the body, and prove the effectiveness and safety of galangin in inhibiting inflammatory response and injury. Methods 120 rats were randomly divided into 6 groups: (Group 1) LPS group; (Group 2) LPS + galangin group; (Group 3) LPS + galangin + GW9662 group; (Group 4) LPS + galangin + DNMT3A siRNA group; (Group 5) LPS + galangin + siRNA negative group; (Group 6) control group. The model of inflammatory lung injury was established by intrathecal instillation of LPS in the first five groups and NS in the control group. SD survival rate was recorded every 24 hours after modeling, lasting for 168 hours. The lung tissues were taken 168 hours after the establishment of the model. The pathological morphology of lung tissue was observed after the staining under the light microscope, and the lung dry/wet weight ratio was calculated after drying. After NS was perfused into lung tissue, the lavage fluid was collected and the levels of IL-6 and TNF-a were measured by ELISA. The contents of PPAR, DNMT3A, phosphorylated p65, and ERK in monocytes were detected by the WB method, and the binding contents of p65 and AP-1 in the promoter regions of IL-6 and TNF-a genes were detected by the Chip-qPCR method. Results Intraperitoneal injection of galangin could inhibit the synthesis of alveolar inflammatory factors (TFs) in the SD model of lung injury induced by LPS, reduce the degree of pathological injury of lung tissue, and improve the survival rate of the SD model. GW9662 can completely reverse the protective effect, while DNMT3A interference can only partially block its protective effect. In addition, galangin could significantly inhibit the LPS-induced expression of p65 and AP-1 in alveolar monocytes and their binding content in the promoter region of inflammatory genes by activating PPAR/DNMT3A pathway. GW9662 could completely reverse the inhibitory effect of galangin. DNMT3A interference could restore the binding content of transcription factors at the promoter of the inflammatory gene but had no significant effect on its synthesis. Conclusion Galangin can interfere with the binding of transcription factors to inflammatory gene promoters through the methylation modification induced by PPAR/DNMT3A pathway, so as to inhibit the synthesis of inflammatory molecules and reverse inflammatory lung injury.
Collapse
|
35
|
Reutersberg B, Düppers P, Menges AL, Schrimpf C, Zimmermann A, Pelisek J. Alterungsbedingte Gefäßveränderungen am Beispiel der Arteria carotis. GEFÄSSCHIRURGIE 2022; 27:231-238. [PMID: 35789693 PMCID: PMC9243795 DOI: 10.1007/s00772-022-00901-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/16/2022] [Indexed: 11/29/2022]
Abstract
Einer der Hauptrisikofaktoren für das Vorliegen einer Karotisstenose und des karotisbedingten Schlaganfalls ist das Lebensalter. Ziel dieses Übersichtsartikels ist die Darstellung des aktuellen Wissensstands über altersbedingte Veränderungen der Gefäße am Beispiel der Karotisstenose. Die Gefäßalterung (vaskuläre Seneszenz) als Abnahme struktureller und funktioneller Eigenschaften der Gefäßwand spielt sich auf verschiedenen Ebenen ab. Auf multizellulärer Ebene kommt es mit zunehmendem Alter hauptsächlich aufgrund von atherosklerotischen Veränderungen der Gefäßwand zu einer Zunahme von Gefäßvolumen und -durchmesser sowie der Intima-Media-Dicke. Auf zellulärer und extrazellulärer Ebene kommt es zur Abnahme von Elastinfasern, glatten Muskelzellen und der Gesamtzellularität sowie zur Zunahme der Lipid‑, Cholesterin- und Kalziumphosphatablagerungen und der Neovaskularisierung. Ursachen der Gefäßalterung auf molekularer Ebene sind insbesondere oxidativer Stress, chronische Entzündungsreaktion, mitochondriale Dysfunktion, epigenetische Veränderungen, Dysregulation der Expression nicht kodierender RNAs (ncRNAs) und die Zunahme der Seneszenz. Der altersbedingte Verlust der Heilungs- und Reparaturfähigkeit des Gewebes macht die Plaques vulnerabler und im Falle der A. carotis anfälliger für ischämische Schlaganfälle. Zunehmende Erkenntnisse über den Einfluss des Alterns auf die Epigenetik und der ncRNAs in atherosklerotischen Plaques kann zukünftig das individuelle Risiko von Patienten genauer quantifizieren und zur Entwicklung zielgerichteter Therapiestrategien beitragen. Weitere Studien sind auf diesem Gebiet jedoch notwendig, um das gesamte Ausmaß der Gefäßalterung und den damit einhergehenden Erkrankungen zu verstehen, damit diesen dann gezielt entgegenwirkt werden kann.
Collapse
Affiliation(s)
- Benedikt Reutersberg
- Klinik für Gefäßchirurgie, Universitätsspital Zürich, Rämistr. 100, 8091 Zürich, Schweiz
| | - Philip Düppers
- Klinik für Gefäßchirurgie, Universitätsspital Zürich, Rämistr. 100, 8091 Zürich, Schweiz
| | - Anna-Leonie Menges
- Klinik für Gefäßchirurgie, Universitätsspital Zürich, Rämistr. 100, 8091 Zürich, Schweiz
| | - Claudia Schrimpf
- Klinik für Gefäßchirurgie, Universitätsspital Zürich, Rämistr. 100, 8091 Zürich, Schweiz
| | - Alexander Zimmermann
- Klinik für Gefäßchirurgie, Universitätsspital Zürich, Rämistr. 100, 8091 Zürich, Schweiz
| | - Jaroslav Pelisek
- Klinik für Gefäßchirurgie, Universitätsspital Zürich, Rämistr. 100, 8091 Zürich, Schweiz
| |
Collapse
|
36
|
Cao YC, Shan SK, Guo B, Li CC, Li FXZ, Zheng MH, Xu QS, Wang Y, Lei LM, Tang KX, Ou-Yang WL, Duan JY, Wu YY, Ullah MHE, Zhou ZA, Xu F, Lin X, Wu F, Liao XB, Yuan LQ. Histone Lysine Methylation Modification and Its Role in Vascular Calcification. Front Endocrinol (Lausanne) 2022; 13:863708. [PMID: 35784574 PMCID: PMC9243330 DOI: 10.3389/fendo.2022.863708] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/06/2022] [Indexed: 01/10/2023] Open
Abstract
Histone methylation is an epigenetic change mediated by histone methyltransferase, and has been connected to the beginning and progression of several diseases. The most common ailments that affect the elderly are cardiovascular and cerebrovascular disorders. They are the leading causes of death, and their incidence is linked to vascular calcification (VC). The key mechanism of VC is the transformation of vascular smooth muscle cells (VSMCs) into osteoblast-like phenotypes, which is a highly adjustable process involving a variety of complex pathophysiological processes, such as metabolic abnormalities, apoptosis, oxidative stress and signalling pathways. Many researchers have investigated the mechanism of VC and related targets for the prevention and treatment of cardiovascular and cerebrovascular diseases. Their findings revealed that histone lysine methylation modification may play a key role in the various stages of VC. As a result, a thorough examination of the role and mechanism of lysine methylation modification in physiological and pathological states is critical, not only for identifying specific molecular markers of VC and new therapeutic targets, but also for directing the development of new related drugs. Finally, we provide this review to discover the association between histone methylation modification and VC, as well as diverse approaches with which to investigate the pathophysiology of VC and prospective treatment possibilities.
Collapse
Affiliation(s)
- Ye-Chi Cao
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Su-Kang Shan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bei Guo
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chang-Chun Li
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Fu-Xing-Zi Li
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ming-Hui Zheng
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qiu-Shuang Xu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Wang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Li-Min Lei
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ke-Xin Tang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Lu Ou-Yang
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Yue Duan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yun-Yun Wu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Muhammad Hasnain Ehsan Ullah
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhi-Ang Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Xu
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao Lin
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Feng Wu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Bo Liao
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Qing Yuan
- National Clinical Research Center for Metabolic Diseases, Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
37
|
Carballo-Perich L, Puigoriol-Illamola D, Bashir S, Terceño M, Silva Y, Gubern-Mérida C, Serena J. Clinical Parameters and Epigenetic Biomarkers of Plaque Vulnerability in Patients with Carotid Stenosis. Int J Mol Sci 2022; 23:5149. [PMID: 35563540 PMCID: PMC9101730 DOI: 10.3390/ijms23095149] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 12/24/2022] Open
Abstract
Atheromatous disease is the first cause of death and dependency in developed countries and carotid artery atherosclerosis is one of the main causes of severe ischaemic strokes. Current management strategies are mainly based on the degree of stenosis and patient selection has limited accuracy. This information could be complemented by the identification of biomarkers of plaque vulnerability, which would permit patients at greater and lesser risk of stroke to be distinguished, thus enabling a better selection of patients for surgical or intensive medical treatment. Although several circulating protein-based biomarkers with significance for both the diagnosis of carotid artery disease and its prognosis have been identified, at present, none have been clinically implemented. This review focuses especially on the most relevant clinical parameters to take into account in routine clinical practice and summarises the most up-to-date data on epigenetic biomarkers of carotid atherosclerosis and plaque vulnerability.
Collapse
Affiliation(s)
- Laia Carballo-Perich
- Cerebrovascular Pathology Research Group, Girona Biomedical Research Institute (IDIBGI), RICORS-ICTUS, Parc Hospitalari Martí I Julià, Edifici M2, 17190 Salt, Spain; (L.C.-P.); (D.P.-I.)
| | - Dolors Puigoriol-Illamola
- Cerebrovascular Pathology Research Group, Girona Biomedical Research Institute (IDIBGI), RICORS-ICTUS, Parc Hospitalari Martí I Julià, Edifici M2, 17190 Salt, Spain; (L.C.-P.); (D.P.-I.)
| | - Saima Bashir
- Cerebrovascular Pathology Research Group, Stroke Unit, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, RICORS-ICTUS, Av. França s/n (7a Planta), 17007 Girona, Spain; (S.B.); (M.T.); (J.S.)
| | - Mikel Terceño
- Cerebrovascular Pathology Research Group, Stroke Unit, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, RICORS-ICTUS, Av. França s/n (7a Planta), 17007 Girona, Spain; (S.B.); (M.T.); (J.S.)
| | - Yolanda Silva
- Cerebrovascular Pathology Research Group, Stroke Unit, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, RICORS-ICTUS, Av. França s/n (7a Planta), 17007 Girona, Spain; (S.B.); (M.T.); (J.S.)
| | - Carme Gubern-Mérida
- Cerebrovascular Pathology Research Group, Girona Biomedical Research Institute (IDIBGI), RICORS-ICTUS, Parc Hospitalari Martí I Julià, Edifici M2, 17190 Salt, Spain; (L.C.-P.); (D.P.-I.)
| | - Joaquín Serena
- Cerebrovascular Pathology Research Group, Stroke Unit, Department of Neurology, Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, RICORS-ICTUS, Av. França s/n (7a Planta), 17007 Girona, Spain; (S.B.); (M.T.); (J.S.)
| |
Collapse
|
38
|
Lin Y, Qiu T, Wei G, Que Y, Wang W, Kong Y, Xie T, Chen X. Role of Histone Post-Translational Modifications in Inflammatory Diseases. Front Immunol 2022; 13:852272. [PMID: 35280995 PMCID: PMC8908311 DOI: 10.3389/fimmu.2022.852272] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Inflammation is a defensive reaction for external stimuli to the human body and generally accompanied by immune responses, which is associated with multiple diseases such as atherosclerosis, type 2 diabetes, Alzheimer’s disease, psoriasis, asthma, chronic lung diseases, inflammatory bowel disease, and multiple virus-associated diseases. Epigenetic mechanisms have been demonstrated to play a key role in the regulation of inflammation. Common epigenetic regulations are DNA methylation, histone modifications, and non-coding RNA expression; among these, histone modifications embrace various post-modifications including acetylation, methylation, phosphorylation, ubiquitination, and ADP ribosylation. This review focuses on the significant role of histone modifications in the progression of inflammatory diseases, providing the potential target for clinical therapy of inflammation-associated diseases.
Collapse
Affiliation(s)
- Yingying Lin
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Ting Qiu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Guifeng Wei
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Yueyue Que
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Wenxin Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yichao Kong
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Xiabin Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, China.,Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
39
|
Yang H, Sun Y, Li Q, Jin F, Dai Y. Diverse Epigenetic Regulations of Macrophages in Atherosclerosis. Front Cardiovasc Med 2022; 9:868788. [PMID: 35425818 PMCID: PMC9001883 DOI: 10.3389/fcvm.2022.868788] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/04/2022] [Indexed: 02/05/2023] Open
Abstract
Emerging research on epigenetics has resulted in many novel discoveries in atherosclerosis (AS), an inflammaging-associated disease characterized by chronic inflammation primarily driven by macrophages. The bulk of evidence has demonstrated the central role of epigenetic machinery in macrophage polarization to pro- (M1-like) or anti-inflammatory (M2-like) phenotype. An increasing number of epigenetic alterations and their modifiers involved in reprogramming macrophages by regulating DNA methylation or histone modifications (e.g., methylation, acetylation, and recently lactylation) have been identified. They may act to determine or skew the direction of macrophage polarization in AS lesions, thereby representing a promising target. Here we describe the current understanding of the epigenetic machinery involving macrophage polarization, to shed light on chronic inflammation-driving onset and progression of inflammaging-associated diseases, using AS as a prototypic example, and discuss the challenge for developing effective therapies targeting the epigenetic modifiers against these diseases, particularly highlighting a potential strategy based on epigenetically-governed repolarization from M1-like to M2-like phenotype.
Collapse
Affiliation(s)
- Hongmei Yang
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
- Department of Critical Care Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yue Sun
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Qingchao Li
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Fengyan Jin
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
40
|
Li RL, Wang LY, Liu S, Duan HX, Zhang Q, Zhang T, Peng W, Huang Y, Wu C. Natural Flavonoids Derived From Fruits Are Potential Agents Against Atherosclerosis. Front Nutr 2022; 9:862277. [PMID: 35399657 PMCID: PMC8987282 DOI: 10.3389/fnut.2022.862277] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis, as a chronic inflammatory response, is one of the main causes of cardiovascular diseases. Atherosclerosis is induced by endothelial cell dysfunction, migration and proliferation of smooth muscle cells, accumulation of foam cells and inflammatory response, resulting in plaque accumulation, narrowing and hardening of the artery wall, and ultimately leading to myocardial infarction or sudden death and other serious consequences. Flavonoid is a kind of natural polyphenol compound widely existing in fruits with various structures, mainly including flavonols, flavones, flavanones, flavanols, anthocyanins, isoflavones, and chalcone, etc. Because of its potential health benefits, it is now used in supplements, cosmetics and medicines, and researchers are increasingly paying attention to its role in atherosclerosis. In this paper, we will focus on several important nodes in the development of atherosclerotic disease, including endothelial cell dysfunction, smooth muscle cell migration and proliferation, foam cell accumulation and inflammatory response. At the same time, through the classification of flavonoids from fruits, the role and potential mechanism of flavonoids in atherosclerosis were reviewed, providing a certain direction for the development of fruit flavonoids in the treatment of atherosclerosis drugs.
Collapse
Affiliation(s)
- Ruo-Lan Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ling-Yu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuqin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hu-Xinyue Duan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qing Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Wei Peng,
| | - Yongliang Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Yongliang Huang,
| | - Chunjie Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chunjie Wu,
| |
Collapse
|
41
|
Yi X, Zhu QX, Wu XL, Tan TT, Jiang XJ. Histone Methylation and Oxidative Stress in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6023710. [PMID: 35340204 PMCID: PMC8942669 DOI: 10.1155/2022/6023710] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 02/17/2022] [Accepted: 03/05/2022] [Indexed: 11/18/2022]
Abstract
Oxidative stress occurs when ROS overproduction overwhelms the elimination ability of antioxidants. Accumulated studies have found that oxidative stress is regulated by histone methylation and plays a critical role in the development and progression of cardiovascular diseases. Targeting the underlying molecular mechanism to alter the interplay of oxidative stress and histone methylation may enable creative and effective therapeutic strategies to be developed against a variety of cardiovascular disorders. Recently, some drugs targeting epigenetic modifiers have been used to treat specific types of cancers. However, the comprehensive signaling pathways bridging oxidative stress and histone methylation need to be deeply explored in the contexts of cardiovascular physiology and pathology before clinical therapies be developed. In the present review, we summarize and update information on the interplay between histone methylation and oxidative stress during the development of cardiovascular diseases such as atherosclerosis, coronary artery disease, pulmonary hypertension, and diabetic macro- and microvascular pathologies.
Collapse
Affiliation(s)
- Xin Yi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Qiu-Xia Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Xing-Liang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| | - Tuan-Tuan Tan
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xue-Jun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Cardiovascular Research Institute, Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Cardiology, Wuhan 430060, China
| |
Collapse
|
42
|
Wang K, Gao H, Zhang Y, Yan H, Si J, Mi X, Xia S, Feng X, Liu D, Kong D, Wang T, Ding D. Highly Bright AIE Nanoparticles by Regulating the Substituent of Rhodanine for Precise Early Detection of Atherosclerosis and Drug Screening. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106994. [PMID: 34921573 DOI: 10.1002/adma.202106994] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 12/15/2021] [Indexed: 06/14/2023]
Abstract
Fluorescent probes capable of precise detection of atherosclerosis (AS) at an early stage and fast assessment of anti-AS drugs in animal level are particularly valuable. Herein, a highly bright aggregation-induced emission (AIE) nanoprobe is introduced by regulating the substituent of rhodanine for early detection of atherosclerotic plaque and screening of anti-AS drugs in a precise, sensitive, and rapid manner. With dicyanomethylene-substituted rhodanine as the electron-withdrawing unit, the AIE luminogen named TPE-T-RCN shows the highest molar extinction coefficient, the largest photoluminescence quantum yield, and the most redshifted absorption/emission spectra simultaneously as compared to the control compounds. The nanoprobes are obtained with an amphiphilic copolymer as the matrix encapsulating TPE-T-RCN molecules, which are further surface functionalized with anti-CD47 antibody for specifically binding to CD47 overexpressed in AS plaques. Such nanoprobes allow efficient recognition of AS plaques at different stages in apolipoprotein E-deficient (apoE-/- ) mice, especially for the recognition of early-stage AS plaques prior to micro-computed tomography (CT) and magnetic resonance imaging (MRI). These features impel to apply the nanoprobes in monitoring the therapeutic effects of anti-AS drugs, providing a powerful tool for anti-AS drug screening. Their potential use in targeted imaging of human carotid plaque is further demonstrated.
Collapse
Affiliation(s)
- Kai Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Heqi Gao
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yuwen Zhang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Hongyu Yan
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Jianghua Si
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Xingyan Mi
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Shuang Xia
- Department of Radiology, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Xuequan Feng
- Department of Neurosurgery, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Dingbin Liu
- College of Chemistry, Research Center for Analytical Sciences, State Key Laboratory of Medicinal Chemical Biology, and Tianjin Key Laboratory of Molecular Recognition and Biosensing, Nankai University, Tianjin, 300071, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
| | - Ting Wang
- Tianjin Key Laboratory of Urban Transport Emission Research, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Dan Ding
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Frontiers Science Center for Cell Responses, and State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin Stomatological Hospital, The Affiliated Stomatological Hospital of Nankai University, Tianjin, 300041, China
| |
Collapse
|
43
|
Sharma R, Sharma S, Thakur A, Singh A, Singh J, Nepali K, Liou JP. The Role of Epigenetic Mechanisms in Autoimmune, Neurodegenerative, Cardiovascular, and Imprinting Disorders. Mini Rev Med Chem 2022; 22:1977-2011. [PMID: 35176978 DOI: 10.2174/1389557522666220217103441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/01/2021] [Accepted: 11/11/2021] [Indexed: 11/22/2022]
Abstract
Epigenetic mutations like aberrant DNA methylation, histone modifications, or RNA silencing are found in a number of human diseases. This review article discusses the epigenetic mechanisms involved in neurodegenerative disorders, cardiovascular disorders, auto-immune disorder, and genomic imprinting disorders. In addition, emerging epigenetic therapeutic strategies for the treatment of such disorders are presented. Medicinal chemistry campaigns highlighting the efforts of the chemists invested towards the rational design of small molecule inhibitors have also been included. Pleasingly, several classes of epigenetic inhibitors, DNMT, HDAC, BET, HAT, and HMT inhibitors along with RNA based therapies have exhibited the potential to emerge as therapeutics in the longer run. It is quite hopeful that epigenetic modulator-based therapies will advance to clinical stage investigations by leaps and bounds.
Collapse
Affiliation(s)
- Ram Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Sachin Sharma
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Arshdeep Singh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jagjeet Singh
- School of Pharmacy, University of Queensland, Brisbane, QLD, Australia.,Department of Pharmacy, Rayat-Bahara Group of Institutes, Hoshiarpur, India
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Jing Ping Liou
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
44
|
Tang H, Zeng Z, Shang C, Li Q, Liu J. Epigenetic Regulation in Pathology of Atherosclerosis: A Novel Perspective. Front Genet 2022; 12:810689. [PMID: 34976029 PMCID: PMC8714670 DOI: 10.3389/fgene.2021.810689] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/02/2021] [Indexed: 12/19/2022] Open
Abstract
Atherosclerosis, characterized by atherosclerotic plaques, is a complex pathological process that involves different cell types and can be seen as a chronic inflammatory disease. In the advanced stage, the ruptured atherosclerotic plaque can induce deadly accidents including ischemic stroke and myocardial infarction. Epigenetics regulation, including DNA methylation, histone modification, and non-coding RNA modification. maintains cellular identity via affecting the cellular transcriptome. The epigenetic modification process, mediating by epigenetic enzymes, is dynamic under various stimuli, which can be reversely altered. Recently, numerous studies have evidenced the close relationship between atherosclerosis and epigenetic regulations in atherosclerosis, providing us with a novel perspective in researching mechanisms and finding novel therapeutic targets of this serious disease. Here, we critically review the recent discoveries between epigenetic regulation mechanisms in atherosclerosis.
Collapse
Affiliation(s)
- Haishuang Tang
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Zhangwei Zeng
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Chenghao Shang
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Qiang Li
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, Shanghai, China
| | - Jianmin Liu
- Department of Neurosurgery, Changhai Hospital, Naval Military Medical University, Shanghai, China
| |
Collapse
|
45
|
Bompada P, Goncalves I, Wu C, Gao R, Sun J, Mir BA, Luan C, Renström E, Groop L, Weng J, Hansson O, Edsfeldt A, De Marinis Y. Epigenome-Wide Histone Acetylation Changes in Peripheral Blood Mononuclear Cells in Patients with Type 2 Diabetes and Atherosclerotic Disease. Biomedicines 2021; 9:biomedicines9121908. [PMID: 34944721 PMCID: PMC8698994 DOI: 10.3390/biomedicines9121908] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022] Open
Abstract
There is emerging evidence of an association between epigenetic modifications, glycemic control and atherosclerosis risk. In this study, we mapped genome-wide epigenetic changes in patients with type 2 diabetes (T2D) and advanced atherosclerotic disease. We performed chromatin immunoprecipitation sequencing (ChIP-seq) using a histone 3 lysine 9 acetylation (H3K9ac) mark in peripheral blood mononuclear cells from patients with atherosclerosis with T2D (n = 8) or without T2D (ND, n = 10). We mapped epigenome changes and identified 23,394 and 13,133 peaks in ND and T2D individuals, respectively. Out of all the peaks, 753 domains near the transcription start site (TSS) were unique to T2D. We found that T2D in atherosclerosis leads to an H3K9ac increase in 118, and loss in 63 genomic regions. Furthermore, we discovered an association between the genomic locations of significant H3K9ac changes with genetic variants identified in previous T2D GWAS. The transcription factor 7-like 2 (TCF7L2) rs7903146, together with several human leukocyte antigen (HLA) variants, were among the domains with the most dramatic changes of H3K9ac enrichments. Pathway analysis revealed multiple activated pathways involved in immunity, including type 1 diabetes. Our results present novel evidence on the interaction between genetics and epigenetics, as well as epigenetic changes related to immunity in patients with T2D and advanced atherosclerotic disease.
Collapse
Affiliation(s)
- Pradeep Bompada
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
| | - Isabel Goncalves
- Cardiovascular Research-Translational Studies, Institution of Clinical Science Malmö, Lund University, 20502 Malmö, Sweden; (I.G.); (J.S.); (A.E.)
- Department of Cardiology, Skåne University Hospital, 20502 Malmö, Sweden
| | - Chuanyan Wu
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
- School of Control Science and Engineering, Shandong University, Jinan 250061, China
- School of Intelligent Engineering, Shandong Management University, Jinan 250100, China
| | - Rui Gao
- School of Control Science and Engineering, Shandong University, Jinan 250061, China
- Correspondence: (R.G.); (Y.D.M.); Tel.: +86-135-0531-8418 (R.G.); +46-760-384-868 (Y.D.M.)
| | - Jiangming Sun
- Cardiovascular Research-Translational Studies, Institution of Clinical Science Malmö, Lund University, 20502 Malmö, Sweden; (I.G.); (J.S.); (A.E.)
| | - Bilal Ahmad Mir
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
| | - Cheng Luan
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
| | - Erik Renström
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
| | - Leif Groop
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
- Finnish Institute for Molecular Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Jianping Weng
- Clinical Research Hospital, Chinese Academy of Sciences, Hefei 230001, China;
- Department of Endocrinology and Metabolism, Division of Life Sciences of Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ola Hansson
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
- Institute for Molecular Medicine Finland (FIMM), Helsinki University, 00290 Helsinki, Finland
| | - Andreas Edsfeldt
- Cardiovascular Research-Translational Studies, Institution of Clinical Science Malmö, Lund University, 20502 Malmö, Sweden; (I.G.); (J.S.); (A.E.)
- Department of Cardiology, Skåne University Hospital, 20502 Malmö, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, 20502 Malmö, Sweden
| | - Yang De Marinis
- Department of Clinical Sciences, Lund University, 20502 Malmö, Sweden; (P.B.); (C.W.); (B.A.M.); (C.L.); (E.R.); (L.G.); (O.H.)
- School of Control Science and Engineering, Shandong University, Jinan 250061, China
- Clinical Research Hospital, Chinese Academy of Sciences, Hefei 230001, China;
- Department of Endocrinology and Metabolism, Division of Life Sciences of Medicine, University of Science and Technology of China, Hefei 230001, China
- Correspondence: (R.G.); (Y.D.M.); Tel.: +86-135-0531-8418 (R.G.); +46-760-384-868 (Y.D.M.)
| |
Collapse
|
46
|
Gao L, Yu W, Song P, Li Q. Non-histone methylation of SET7/9 and its biological functions. Recent Pat Anticancer Drug Discov 2021; 17:231-243. [PMID: 34856916 DOI: 10.2174/1574892816666211202160041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND (su(var)-3-9,enhancer-of-zeste,trithorax) domain-containing protein 7/9 (SET7/9) is a member of the protein lysine methyltransferases (PLMTs or PKMTs) family. It contains a SET domain. Recent studies demonstrate that SET7/9 methylates both lysine 4 of histone 3 (H3-K4) and lysine(s) of non-histone proteins, including transcription factors, tumor suppressors, and membrane-associated receptors. OBJECTIVE This article mainly reviews the non-histone methylation effects of SET7/9 and its functions in tumorigenesis and development. METHODS PubMed was screened for this information. RESULTS SET7/9 plays a key regulatory role in various biological processes such as cell proliferation, transcription regulation, cell cycle, protein stability, cardiac morphogenesis, and development. In addition, SET7/9 is involved in the pathogenesis of hair loss, breast cancer progression, human carotid plaque atherosclerosis, chronic kidney disease, diabetes, obesity, ovarian cancer, prostate cancer, hepatocellular carcinoma, and pulmonary fibrosis. CONCLUSION SET7/9 is an important methyltransferase, which can catalyze the methylation of a variety of proteins. Its substrates are closely related to the occurrence and development of tumors.
Collapse
Affiliation(s)
- Lili Gao
- Department of Pathology, Pudong New Area People's Hospital, Shanghai 201299. China
| | - Weiping Yu
- Department of Pathophysiology, Medical school of Southeast University, Nanjing 210009, Jiangsu. China
| | - Peng Song
- Department of Pathology, Pudong New Area People's Hospital, Shanghai 201299. China
| | - Qing Li
- Department of Pathology, Pudong New Area People's Hospital, Shanghai 201299. China
| |
Collapse
|
47
|
Castro R, Whalen CA, Gullette S, Mattie FJ, Florindo C, Heil SG, Huang NK, Neuberger T, Ross AC. A Hypomethylating Ketogenic Diet in Apolipoprotein E-Deficient Mice: A Pilot Study on Vascular Effects and Specific Epigenetic Changes. Nutrients 2021; 13:nu13103576. [PMID: 34684577 PMCID: PMC8537671 DOI: 10.3390/nu13103576] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 09/27/2021] [Accepted: 10/02/2021] [Indexed: 12/20/2022] Open
Abstract
Hyperhomocysteneinemia (HHcy) is common in the general population and is a risk factor for atherosclerosis by mechanisms that are still elusive. A hypomethylated status of epigenetically relevant targets may contribute to the vascular toxicity associated with HHcy. Ketogenic diets (KD) are diets with a severely restricted amount of carbohydrates that are being widely used, mainly for weight-loss purposes. However, studies associating nutritional ketosis and HHcy are lacking. This pilot study investigates the effects of mild HHcy induced by nutritional manipulation of the methionine metabolism in the absence of dietary carbohydrates on disease progression and specific epigenetic changes in the apolipoprotein-E deficient (apoE-/-) mouse model. ApoE-/- mice were either fed a KD, a diet with the same macronutrient composition but low in methyl donors (low methyl KD, LMKD), or control diet. After 4, 8 or 12 weeks plasma was collected for the quantification of: (1) nutritional ketosis, (i.e., the ketone body beta-hydroxybutyrate using a colorimetric assay); (2) homocysteine by HPLC; (3) the methylating potential S-adenosylmethionine to S-adenosylhomocysteine ratio (AdoHcy/AdoMet) by LC-MS/MS; and (4) the inflammatory cytokine monocyte chemoattractant protein 1 (MCP1) by ELISA. After 12 weeks, aortas were collected to assess: (1) the vascular AdoHcy/AdoMet ratio; (2) the volume of atherosclerotic lesions by high-field magnetic resonance imaging (14T-MRI); and (3) the content of specific epigenetic tags (H3K27me3 and H3K27ac) by immunofluorescence. The results confirmed the presence of nutritional ketosis in KD and LMKD mice but not in the control mice. As expected, mild HHcy was only detected in the LMKD-fed mice. Significantly decreased MCP1 plasma levels and plaque burden were observed in control mice versus the other two groups, together with an increased content of one of the investigated epigenetic tags (H3K27me3) but not of the other (H3K27ac). Moreover, we are unable to detect any significant differences at the p < 0.05 level for MCP1 plasma levels, vascular AdoMet:AdoHcy ratio levels, plaque burden, and specific epigenetic content between the latter two groups. Nevertheless, the systemic methylating index was significantly decreased in LMKD mice versus the other two groups, reinforcing the possibility that the levels of accumulated homocysteine were insufficient to affect vascular transmethylation reactions. Further studies addressing nutritional ketosis in the presence of mild HHcy should use a higher number of animals and are warranted to confirm these preliminary observations.
Collapse
Affiliation(s)
- Rita Castro
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (C.A.W.); (F.J.M.); (N.K.H.); (A.C.R.)
- Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal;
- Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Correspondence: ; Tel.: +1-814-865-2938
| | - Courtney A. Whalen
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (C.A.W.); (F.J.M.); (N.K.H.); (A.C.R.)
| | - Sean Gullette
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (S.G.); (T.N.)
| | - Floyd J. Mattie
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (C.A.W.); (F.J.M.); (N.K.H.); (A.C.R.)
| | - Cristina Florindo
- Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal;
| | - Sandra G. Heil
- Medical Center Rotterdam, Department of Clinical Chemistry, Erasmus MC University, 3015 GD Rotterdam, The Netherlands;
| | - Neil K. Huang
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (C.A.W.); (F.J.M.); (N.K.H.); (A.C.R.)
- Jean Mayer USDA Human Nutrition Research Center on Aging, Cardiovascular Nutrition Laboratory, Tufts University, Boston, MA 02111, USA
| | - Thomas Neuberger
- The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (S.G.); (T.N.)
- Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - A. Catharine Ross
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA; (C.A.W.); (F.J.M.); (N.K.H.); (A.C.R.)
| |
Collapse
|
48
|
Gharipour M, Mani A, Amini Baghbahadorani M, de Souza Cardoso CK, Jahanfar S, Sarrafzadegan N, de Oliveira C, Silveira EA. How Are Epigenetic Modifications Related to Cardiovascular Disease in Older Adults? Int J Mol Sci 2021; 22:9949. [PMID: 34576113 PMCID: PMC8470616 DOI: 10.3390/ijms22189949] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/16/2022] Open
Abstract
The rate of aging has increased globally during recent decades and has led to a rising burden of age-related diseases such as cardiovascular disease (CVD). At the molecular level, epigenetic modifications have been shown recently to alter gene expression during the life course and impair cellular function. In this regard, several CVD risk factors, such as lifestyle and environmental factors, have emerged as key factors in epigenetic modifications within the cardiovascular system. In this study, we attempted to summarized recent evidence related to epigenetic modification, inflammation response, and CVD in older adults as well as the effect of lifestyle modification as a preventive strategy in this age group. Recent evidence showed that lifestyle and environmental factors may affect epigenetic mechanisms, such as DNA methylation, histone acetylation, and miRNA expression. Several substances or nutrients such as selenium, magnesium, curcumin, and caffeine (present in coffee and some teas) could regulate epigenetics. Similarly, physical inactivity, alcohol consumption, air pollutants, psychological stress, and shift working are well-known modifiers of epigenetic patterns. Understanding the exact ways that lifestyle and environmental factors could affect the expression of genes could help to influence the time of incidence and severity of aging-associated diseases. This review highlighted that a healthy lifestyle throughout the life course, such as a healthy diet rich in fibers, vitamins, and essential elements, and specific fatty acids, adequate physical activity and sleep, smoking cessation, and stress control, could be useful tools in preventing epigenetic changes that lead to impaired cardiovascular function.
Collapse
Affiliation(s)
- Mojgan Gharipour
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
| | - Arya Mani
- Cardiovascular Research Center, Department of Internal Medicine, and Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA;
| | - Mona Amini Baghbahadorani
- Interventional Cardiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
| | - Camila Kellen de Souza Cardoso
- School of Social Sciences and Health, Nutrition Course, Pontifical Catholic University of Goias, Goiânia 74605-010, Brazil;
| | - Shayesteh Jahanfar
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MI 02111, USA;
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 8158388994, Iran;
- Faculty of Medicine, School of Population and Public Health, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Cesar de Oliveira
- Department of Epidemiology & Public Health, Institute of Epidemiology & Health Care, University College London, London WC1E 6BT, UK;
| | - Erika Aparecida Silveira
- Department of Epidemiology & Public Health, Institute of Epidemiology & Health Care, University College London, London WC1E 6BT, UK;
- Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Goiás, Goiânia 74690-900, Brazil
| |
Collapse
|
49
|
Vanchin B, Sol M, Gjaltema RAF, Brinker M, Kiers B, Pereira AC, Harmsen MC, Moonen JRAJ, Krenning G. Reciprocal regulation of endothelial-mesenchymal transition by MAPK7 and EZH2 in intimal hyperplasia and coronary artery disease. Sci Rep 2021; 11:17764. [PMID: 34493753 PMCID: PMC8423795 DOI: 10.1038/s41598-021-97127-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 08/04/2021] [Indexed: 01/02/2023] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is a form of endothelial dysfunction wherein endothelial cells acquire a mesenchymal phenotype and lose endothelial functions, which contributes to the pathogenesis of intimal hyperplasia and atherosclerosis. The mitogen activated protein kinase 7 (MAPK7) inhibits EndMT and decreases the expression of the histone methyltransferase Enhancer-of-Zeste homologue 2 (EZH2), thereby maintaining endothelial quiescence. EZH2 is the catalytic subunit of the Polycomb Repressive Complex 2 that methylates lysine 27 on histone 3 (H3K27me3). It is elusive how the crosstalk between MAPK7 and EZH2 is regulated in the endothelium and if the balance between MAPK7 and EZH2 is disturbed in vascular disease. In human coronary artery disease, we assessed the expression levels of MAPK7 and EZH2 and found that with increasing intima/media thickness ratio, MAPK7 expression decreased, whereas EZH2 expression increased. In vitro, MAPK7 activation decreased EZH2 expression, whereas endothelial cells deficient of EZH2 had increased MAPK7 activity. MAPK7 activation results in increased expression of microRNA (miR)-101, a repressor of EZH2. This loss of EZH2 in turn results in the increased expression of the miR-200 family, culminating in decreased expression of the dual-specificity phosphatases 1 and 6 who may repress MAPK7 activity. Transfection of endothelial cells with miR-200 family members decreased the endothelial sensitivity to TGFβ1-induced EndMT. In endothelial cells there is reciprocity between MAPK7 signaling and EZH2 expression and disturbances in this reciprocal signaling associate with the induction of EndMT and severity of human coronary artery disease.
Collapse
Affiliation(s)
- Byambasuren Vanchin
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ, Groningen, The Netherlands.,Department of Cardiology, School of Medicine, Mongolian National University of Medical Sciences, Jamyan St 3, Ulaanbaatar, 14210, Mongolia
| | - Marloes Sol
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ, Groningen, The Netherlands
| | - Rutger A F Gjaltema
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ, Groningen, The Netherlands
| | - Marja Brinker
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ, Groningen, The Netherlands
| | - Bianca Kiers
- Laboratory of Genetics and Molecular Cardiology (LIM13), Heart Institute (InCor), University of São Paulo, Avenida Dr. Eneas C. Aguiar 44, São Paulo, SP, 05403-000, Brazil
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology (LIM13), Heart Institute (InCor), University of São Paulo, Avenida Dr. Eneas C. Aguiar 44, São Paulo, SP, 05403-000, Brazil
| | - Martin C Harmsen
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ, Groningen, The Netherlands
| | - Jan-Renier A J Moonen
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ, Groningen, The Netherlands.,Department of Pediatric Cardiology, Center for Congenital Heart Diseases, Beatrix Children's Hospital, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (CA40), 9713GZ, Groningen, The Netherlands
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1 (EA11), 9713GZ, Groningen, The Netherlands.
| |
Collapse
|
50
|
Jin F, Li J, Guo J, Doeppner TR, Hermann DM, Yao G, Dai Y. Targeting epigenetic modifiers to reprogramme macrophages in non-resolving inflammation-driven atherosclerosis. EUROPEAN HEART JOURNAL OPEN 2021; 1:oeab022. [PMID: 35919269 PMCID: PMC9241575 DOI: 10.1093/ehjopen/oeab022] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/28/2021] [Accepted: 08/14/2021] [Indexed: 12/14/2022]
Abstract
Epigenomic and epigenetic research has been providing several new insights into a variety of diseases caused by non-resolving inflammation, including cardiovascular diseases. Atherosclerosis (AS) has long been recognized as a chronic inflammatory disease of the arterial walls, characterized by local persistent and stepwise accelerating inflammation without resolution, also known as uncontrolled inflammation. The pathogenesis of AS is driven primarily by highly plastic macrophages via their polarization to pro- or anti-inflammatory phenotypes as well as other novel subtypes recently identified by single-cell sequencing. Although emerging evidence has indicated the key role of the epigenetic machinery in the regulation of macrophage plasticity, the investigation of epigenetic alterations and modifiers in AS and related inflammation is still in its infancy. An increasing number of the epigenetic modifiers (e.g. TET2, DNMT3A, HDAC3, HDAC9, JMJD3, KDM4A) have been identified in epigenetic remodelling of macrophages through DNA methylation or histone modifications (e.g. methylation, acetylation, and recently lactylation) in inflammation. These or many unexplored modifiers function to determine or switch the direction of macrophage polarization via transcriptional reprogramming of gene expression and intracellular metabolic rewiring upon microenvironmental cues, thereby representing a promising target for anti-inflammatory therapy in AS. Here, we review up-to-date findings involving the epigenetic regulation of macrophages to shed light on the mechanism of uncontrolled inflammation during AS onset and progression. We also discuss current challenges for developing an effective and safe anti-AS therapy that targets the epigenetic modifiers and propose a potential anti-inflammatory strategy that repolarizes macrophages from pro- to anti-inflammatory phenotypes.
Collapse
Affiliation(s)
- Fengyan Jin
- Department of Hematology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun, Jilin 130012, China
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, 1 Dong Dan Dahua Road, Dong Cheng District, Beijing 100730, China
| | - Jianfeng Guo
- School of Pharmaceutical Sciences, Jilin University, 1163 Xinmin Street, Changchun 130021, Jilin, China
| | - Thorsten R Doeppner
- Department of Neurology, University of Göttingen Medical School, Robert-Koch-Str. 40 37075, Göttingen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Gang Yao
- Department of Neurology, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun, Jilin 130041, China
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, Institute of Translational Medicine, The First Hospital of Jilin University, 519 Dong Min Zhu Street, Changchun, Jilin 130061, China
| |
Collapse
|