1
|
Ciantar J, Marttila S, Rajić S, Kostiniuk D, Mishra PP, Lyytikäinen LP, Mononen N, Kleber ME, März W, Kähönen M, Raitakari O, Lehtimäki T, Raitoharju E. Identification and functional characterisation of DNA methylation differences between East- and West-originating Finns. Epigenetics 2024; 19:2397297. [PMID: 39217505 PMCID: PMC11382697 DOI: 10.1080/15592294.2024.2397297] [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: 04/29/2024] [Revised: 08/14/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Eastern and Western Finns show a striking difference in coronary heart disease-related mortality; genetics is a known contributor for this discrepancy. Here, we discuss the potential role of DNA methylation in mediating the discrepancy in cardiometabolic disease-risk phenotypes between the sub-populations. We used data from the Young Finns Study (n = 969) to compare the genome-wide DNA methylation levels of East- and West-originating Finns. We identified 21 differentially methylated loci (FDR < 0.05; Δβ >2.5%) and 7 regions (smoothed FDR < 0.05; CpGs ≥ 5). Methylation at all loci and regions associates with genetic variants (p < 5 × 10-8). Independently of genetics, methylation at 11 loci and 4 regions associates with transcript expression, including genes encoding zinc finger proteins. Similarly, methylation at 5 loci and 4 regions associates with cardiometabolic disease-risk phenotypes including triglycerides, glucose, cholesterol, as well as insulin treatment. This analysis was also performed in LURIC (n = 2371), a German cardiovascular patient cohort, and results replicated for the association of methylation at cg26740318 and DMR_11p15 with diabetes-related phenotypes and methylation at DMR_22q13 with triglyceride levels. Our results indicate that DNA methylation differences between East and West Finns may have a functional role in mediating the cardiometabolic disease discrepancy between the sub-populations.
Collapse
Affiliation(s)
- Joanna Ciantar
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Saara Marttila
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Gerontology Research Center, Tampere University, Tampere, Finland
- Tays Research Services, Wellbeing Services County of Pirkanmaa, Tampere University Hospital, Tampere, Finland
| | - Sonja Rajić
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Daria Kostiniuk
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Nina Mononen
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Marcus E Kleber
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany
- SYNLAB MVZ Humangenetik Mannheim, Mannheim, Germany
| | - Winfried März
- Vth Department of Medicine (Nephrology, Hypertensiology, Endocrinology, Diabetology, Rheumatology), Medical Faculty of Mannheim, Heidelberg University, Mannheim, Germany
- Synlab Academy, SYNLAB Holding Deutschland GmbH, Mannheim, Germany
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Tays Research Services, Fimlab Laboratories, and Finnish Cardiovascular Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Emma Raitoharju
- Molecular Epidemiology (MOLE), Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
| |
Collapse
|
2
|
He R, Bi H, He J, Luo Y, Li X, Li Q, Huang R, Tan L. Thyroid hormones and oxidative stress moderated the association between urinary phthalate metabolites and cardiovascular risk factors. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124927. [PMID: 39265773 DOI: 10.1016/j.envpol.2024.124927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/25/2024] [Accepted: 09/07/2024] [Indexed: 09/14/2024]
Abstract
While previous studies suggested that phthalate exposure poses a risk to cardiovascular health, the results are mixed and indicated variability based on population characteristics and health outcomes assessed. Research that simultaneously investigates the association between urinary phthalate metabolites and multiple cardiovascular risk factors within a single study is relatively scarce. This study assessed human exposure to phthalates by determining urinary metabolite concentrations, and applied multiple statistical techniques to systematically evaluate the individual dose-response relationships and joint effects of phthalate exposure on blood lipids, blood pressure, and fasting blood glucose. The results revealed significant negative associations between urinary phthalate metabolites and low-density lipoprotein cholesterol, triglycerides, total cholesterol, diastolic blood pressure, systolic blood pressure, and fasting blood glucose. Significant nonlinear associations were obtained between specific individual metabolites and diastolic blood pressure. The oxidative stress biomarker 8-hydroxydeoxyguanosine levels in urine and thyroid hormone levels in paired serum were measured simultaneously. Then, we examined the indirect roles of thyroid hormones and oxidative stress in the association between urinary phthalate metabolites and cardiovascular risk factors by mediation and moderation analysis. While the mediation effect was not statistically significant, the negative associations of urinary phthalate metabolites with fasting blood glucose, triglyceride, and lipoprotein cholesterol were statistically significant at lower levels of thyroid hormones by moderation analysis. The association was also significant under certain levels of oxidative stress. The results demonstrated that phthalate exposure is associated with several cardiovascular risk factors, and maintaining appropriate oxidative stress levels and ensuring sufficient thyroid hormone levels may attenuate these associations.
Collapse
Affiliation(s)
- Rong He
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, China
| | - Hua Bi
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, China
| | - Jia He
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, China
| | - Yangxu Luo
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, China
| | - Xiaotong Li
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, China
| | - Qin Li
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, China
| | - Rende Huang
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, China
| | - Lei Tan
- Guangzhou Center for Disease Control and Prevention, Guangzhou, 510440, China.
| |
Collapse
|
3
|
Shi B, Wang J, Zhang J, Li J, Hao Y, Lin X, Zhao R. Dapagliflozin Suppresses High Glucose-Induced Proliferation, Oxidative Stress, and Fibrosis by Reducing Mettl3-Induced m6A Modification in Marcks mRNA. Cardiovasc Toxicol 2024:10.1007/s12012-024-09945-3. [PMID: 39560681 DOI: 10.1007/s12012-024-09945-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
Diabetic cardiomyopathy (DCM) is a common and severe complication of Diabetes mellitus (DM). Dapagliflozin (DAPA) is an oral anti-diabetic drug worldwide for the treatment of type 2 DM. However, the action and mechanism of DAPA in cardiac fibrosis during DCM remain vague. Primary cardiac fibroblasts (CFs) were incubated with high glucose (HG) in vitro. Cell proliferation was detected by MTT and EdU assays. Oxidative stress was evaluated by determining the production of reactive oxygen species and malondialdehyde. Cell fibrosis was assessed by detecting fibrosis-related proteins by western blotting. Levels of Mettl3 (Methyltransferase 3) and Marcks (myristoylated alanine-rich C kinase substrate) were measured using qRT-PCR and western blotting. The m6A modification profile was determined by methylated RNA immunoprecipitation assay and the interaction between Mettl3 and Marcks was verified using dual-luciferase reporter and RIP assays. DAPA treatment alleviated HG-induced proliferation, oxidative stress, and fibrosis in CFs. HG promoted the expression of Mettl3 in CFs. Knockdown of Mettl3 reversed HG-induced proliferation, oxidative stress, and fibrosis in CFs; moreover, forced expression of Mettl3 abolished the protective effects of DAPA on CFs under HG condition. Mechanistically, Mettl3 interacted with Marcks in CFs and induced Marcks mRNA m6A modification. HG induced high expression of Marcks in CFs. The overexpression of Marcks could counteract DAPA or Mettl3 knockdown-evoked inhibitory effects on CF proliferation, oxidative stress, and fibrosis under HG condition. Dapagliflozin suppressed HG-induced proliferation, oxidative stress, and fibrosis by reducing Mettl3-induced m6A modification in Marcks mRNA.
Collapse
Affiliation(s)
- Binhao Shi
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
- Department of Cardiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
| | - Jianfei Wang
- Department of Cardiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
| | - Jing Zhang
- Anhui Province Key Laboratory of Occupational Health, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
| | - Ji Li
- Department of Cardiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
- Graduate School of Bengbu Medical University, Bengbu, 233030, Anhui, China
| | - Yancheng Hao
- Department of Cardiology, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
- Graduate School of Bengbu Medical University, Bengbu, 233030, Anhui, China
| | - Xianhe Lin
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Ren Zhao
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
| |
Collapse
|
4
|
Ataollahi Eshkoor S, Fanijavadi S. Dysbiosis-epigenetics-immune system interaction and ageing health problems. J Med Microbiol 2024; 73. [PMID: 39606883 DOI: 10.1099/jmm.0.001921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Background. The growing interest in microbiota-epigenetics-immune system research stems from the understanding that microbiota, a group of micro-organisms colonized in the human body, can influence the gene expression through epigenetic mechanisms and interaction with the immune system. Epigenetics refers to changes in gene activity that are not caused by the alteration in the DNA sequence itself.Discussion. The clinical significance of this research lies in the potential to develop new therapies for diseases linked to the imbalance of these microbial species (dysbiosis), such as cancer and neurodegenerative diseases. The intricate interaction between microbiota and epigenetics involves the production of metabolites and signalling molecules that can impact our health by influencing immune responses, metabolism and inflammation. Understanding these interactions could lead to novel therapeutic strategies targeting microbiota-epigenetic pathways to improve health outcomes.Conclusion. In this context, we aim to review and emphasize the current knowledge and key concepts that link the microbiota to epigenetics and immune system function, exploring their relevance to the development and maintenance of homeostasis and susceptibility to different diseases later in life. We aim to elucidate key concepts concerning the interactions and potential effects among the human gut microbiota, epigenetics, the immune system and ageing diseases linked to dysbiosis.
Collapse
|
5
|
Arbildo-Vega HI, Cruzado-Oliva FH, Coronel-Zubiate FT, Meza-Málaga JM, Luján-Valencia SA, Luján-Urviola E, Echevarria-Goche A, Farje-Gallardo CA, Castillo-Cornock TB, Serquen-Olano K, Padilla-Cáceres T, Caballero-Apaza L, Aguirre-Ipenza R. Periodontal disease and cardiovascular disease: umbrella review. BMC Oral Health 2024; 24:1308. [PMID: 39468505 PMCID: PMC11520879 DOI: 10.1186/s12903-024-04907-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 09/12/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Periodontal disease (PD) is an infectious and inflammatory condition that affects the tissues surrounding and supporting the teeth. It has been suggested that PD may be associated with cardiovascular disease (CVD), one of the leading causes of mortality worldwide. Our study aimed to investigate the association between PD and CVD through an umbrella review. METHODS A comprehensive search was conducted until April 2024 across various electronic databases, including PubMed, Cochrane Library, Scopus, SciELO, Web of Science, Google Scholar, ProQuest Dissertations and Theses, and OpenGrey. Systematic reviews with or without meta-analysis were considered for inclusion, without any limitations on time or language, provided they examined primary studies linking PD with CVD. The AMSTAR-2 tool was employed to assess the quality and overall confidence of the included studies. RESULTS After the initial search, a total of 516 articles were identified. Following the application of selection criteria, 41 articles remained for further consideration. All these studies indicated an association between PD and CVD, with odds ratios and risk ratios ranging from 1.22 to 4.42 and 1.14 to 2.88, respectively. CONCLUSIONS Systematic reviews with high overall confidence support the association between PD, tooth loss, and cardiovascular diseases. However, it is crucial to interpret these results with caution due to methodological limitations. The potential public health relevance justifies preventive and corrective oral health strategies. Additionally, the need for rigorous future research is highlighted to strengthen the evidence and guide effective public health strategies.
Collapse
Affiliation(s)
- Heber Isac Arbildo-Vega
- Faculty of Dentistry, Dentistry School, Universidad de San Martín de Porres, Chiclayo, 14012, Perú
- Faculty of Human Medicine, Human Medicine School, Universidad de San Martín de Porres, Chiclayo, 14012, Perú
| | - Fredy Hugo Cruzado-Oliva
- Faculty of Stomatology, Stomatology School, Universidad Nacional de Trujillo, Trujillo, 13001, Perú
| | - Franz Tito Coronel-Zubiate
- Faculty of Health Sciences, Stomatology School, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, 01001, Perú
| | - Joan Manuel Meza-Málaga
- Faculty of Medicine, Medicine School, Universidad Católica de Santa María, Arequipa, 04013, Perú
- Postgraduate School, Universidad Católica de Santa María, Arequipa, 04013, Perú
| | - Sara Antonieta Luján-Valencia
- Postgraduate School, Universidad Católica de Santa María, Arequipa, 04013, Perú
- Faculty of Dentistry, Dentistry School, Universidad Católica de Santa María, Arequipa, 04013, Perú
| | - Eduardo Luján-Urviola
- Faculty of Dentistry, Universidad Andina Néstor Cáceres Velásquez, Juliaca, 21104, Perú
| | - Adriana Echevarria-Goche
- Faculty of Stomatology, Universidad Peruana Cayetano Heredia, Lima, 15102, Perú
- Department of Dentistry, Dentistry School, Universidad Norbert Wiener, Lima, 15046, Perú
| | - Carlos Alberto Farje-Gallardo
- Faculty of Health Sciences, Stomatology School, Universidad Nacional Toribio Rodríguez de Mendoza de Amazonas, Chachapoyas, 01001, Perú
| | - Tania Belú Castillo-Cornock
- Faculty of Dentistry, Dentistry School, Universidad de San Martín de Porres, Chiclayo, 14012, Perú
- Faculty of Health Sciences, Stomatology School, Universidad Señor de Sipán, Chiclayo, 14000, Perú
| | - Katherine Serquen-Olano
- Faculty of Dentistry, Dentistry School, Universidad de San Martín de Porres, Chiclayo, 14012, Perú
- Faculty of Health Sciences, Stomatology School, Universidad Señor de Sipán, Chiclayo, 14000, Perú
| | - Tania Padilla-Cáceres
- Department of General Dentistry, Dentistry School, Universidad Nacional del Altiplano, Puno, 21001, Perú
- Research Institute in Environmental Sciences, Health and Biodiversity - IICASB, Universidad Nacional del Altiplano, Puno, 21001, Perú
| | - Luz Caballero-Apaza
- Department of Nursing, School of Nursing, Universidad Nacional del Altiplano, Puno, 21001, Perú
| | | |
Collapse
|
6
|
Zeng L, Liang Y, Zhou R, Yang W, Chen K, He B, Qiu Y, Liu L, Zhou D, Xiao Z, Liang H, Zhang B, Li R, Yu L, Yi M, Lin X. PD-1/PD-L1 and coronary heart disease: a mendelian randomization study. Front Cardiovasc Med 2024; 11:1424770. [PMID: 39494235 PMCID: PMC11527656 DOI: 10.3389/fcvm.2024.1424770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
Introduction It has been found that programmed cell death protein-1 (PD-1) or its ligand PD-L1 may play an important role in the onset and progression of coronary heart disease (CHD). Thus, we conducted this mendelian randomization analysis (MR) to estimate the causal relationship between PD-1/PD-L1 and 5 specific CHDs (chronic ischemic heart disease, acute myocardial infarction, angina pectoris, coronary atherosclerosis, and unstable angina pectoris), complemented by gene set enrichment analysis (GSEA) for further validation. Methods Publicly available summary-level data were attained from the UK Biobank with genetic instruments obtained from the largest available, nonoverlapping genome-wide association studies (GWAS). Our analysis involved various approaches including inverse variance-weighted meta-analysis, alternative techniques like weighted median, MR-Egger, MR-multipotency residuals and outliers detection (PRESSO), along with multiple sensitivity assessments such as MR-Egger intercept test, Cochran's Q test, and leave-one-out sensitivity analysis to evaluate and exclude any anomalies. Results Gene expression profile (GSE71226) was obtained from Gene Expression Omnibus (GEO) database for GSEA. IVW analysis showed a causal association between PD-1 and chronic ischemic heart disease (OR, 0.997; 95%CI, 0.995-0.999; P, 0.009), chronic ischemic heart disease and PD-1 (beta, -3.1; 95%CI, -6.017 to -0.183; P, 0.037), chronic ischemic heart disease and PD-L1 (beta, -3.269; 95%CI, -6.197 to -0.341; P, 0.029). No significant causal relationship was found between PD-1/PD-L1 and other 4 CHDs. The accuracy and robustness of these findings were confirmed by sensitivity tests. GSEA found that the KEGG pathway and related core genes of "PD-L1 expression and PD-1 checkpoint pathway in cancer" pathway were downregulated in CHD. Discussion This study provided evidence of a bidirectional causal relationship between PD-1 and chronic ischemic heart disease and a protective association between chronic ischemic heart disease and PD-L1.
Collapse
Affiliation(s)
- Liangjia Zeng
- The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
| | - Yinglan Liang
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
- Department of Anesthesiology, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
| | - Ruoyun Zhou
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
| | - Wenting Yang
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
- Department of Medical Imageology, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
| | - Kexin Chen
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
| | - Baixin He
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
| | - Yuqing Qiu
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
| | - Linglong Liu
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
- Department of Anesthesiology, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
| | - Deyang Zhou
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
- Department of Anesthesiology, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
| | - Zhaolin Xiao
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
| | - Haowen Liang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
| | - Binghua Zhang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
| | - Renyu Li
- Medical Exploration and Translation Team, Cardiovascular Medicine and Cardio-Oncology Group, Guangzhou, China
- Department of Psychiatry, Clinical Medical School, Guangzhou Medical University, Guangzhou, China
| | - Lihong Yu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Min Yi
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaozhen Lin
- Department of Cardiology, Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
7
|
Du Q, Zhang Z, Yang W, Zhou X, Zhou N, Wu C, Bao J. CBGDA: a manually curated resource for gene-disease associations based on genome-wide CRISPR. Database (Oxford) 2024; 2024:baae077. [PMID: 39213392 PMCID: PMC11363955 DOI: 10.1093/database/baae077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/16/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
The field of understanding the association between genes and diseases is rapidly expanding, making it challenging for researchers to keep up with the influx of new publications and genetic datasets. Fortunately, there are now several regularly updated databases available that focus on cataloging gene-disease relationships. The development of the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 system has revolutionized the field of gene editing, providing a highly efficient, accurate, and reliable method for exploring gene-disease associations. However, currently, there is no resource specifically dedicated to collecting and integrating the latest experimentally supported gene-disease association data derived from genome-wide CRISPR screening. To address this gap, we have developed the CRISPR-Based Gene-Disease Associations (CBGDA) database, which includes over 200 manually curated gene-disease association data derived from genome-wide CRISPR screening studies. Through CBGDA, users can explore gene-disease association data derived from genome-wide CRISPR screening, gaining insights into the expression patterns of genes in different diseases, associated chemical data, and variant information. This provides a novel perspective on understanding the associations between genes and diseases. What is more, CBGDA integrates data from several other databases and resources, enhancing its comprehensiveness and utility. In summary, CBGDA offers a fresh perspective and comprehensive insights into the research on gene-disease associations. It fills the gap by providing a dedicated resource for accessing up-to-date, experimentally supported gene-disease association data derived from genome-wide CRISPR screening. Database URL: http://cbgda.zhounan.org/main.
Collapse
Affiliation(s)
- Qingsong Du
- Key Laboratory of the State Ministry of Education for Bio-Resources and Ecologic Environment, College of Life Sciences, Sichuan University, 29 Wangjiang Rd, Chengdu 610064, China
| | - Zhiyu Zhang
- Key Laboratory of the State Ministry of Education for Bio-Resources and Ecologic Environment, College of Life Sciences, Sichuan University, 29 Wangjiang Rd, Chengdu 610064, China
| | - Wanyi Yang
- Key Laboratory of the State Ministry of Education for Bio-Resources and Ecologic Environment, College of Life Sciences, Sichuan University, 29 Wangjiang Rd, Chengdu 610064, China
| | - Xunyu Zhou
- Key Laboratory of the State Ministry of Education for Bio-Resources and Ecologic Environment, College of Life Sciences, Sichuan University, 29 Wangjiang Rd, Chengdu 610064, China
| | - Nan Zhou
- Research Center, The Affiliated Brain Hospital, Guangzhou Medical University, 36 Mingxin Rd, Guangzhou 510000, China
| | - Chuanfang Wu
- Key Laboratory of the State Ministry of Education for Bio-Resources and Ecologic Environment, College of Life Sciences, Sichuan University, 29 Wangjiang Rd, Chengdu 610064, China
| | - Jinku Bao
- Key Laboratory of the State Ministry of Education for Bio-Resources and Ecologic Environment, College of Life Sciences, Sichuan University, 29 Wangjiang Rd, Chengdu 610064, China
| |
Collapse
|
8
|
Yang X, Huang Y, Tang D, Yue L. Identification of key genes associated with acute myocardial infarction using WGCNA and two-sample mendelian randomization study. PLoS One 2024; 19:e0305532. [PMID: 39024234 PMCID: PMC11257238 DOI: 10.1371/journal.pone.0305532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 06/02/2024] [Indexed: 07/20/2024] Open
Abstract
OBJECTIVE Acute myocardial infarction (AMI) is a severe condition with high morbidity and mortality rates. This study aimed to identify hub genes potentially associated with AMI and assess their clinical utility in predicting AMI occurrence. METHODS Gene microarray data were obtained from the Gene Expression Omnibus (GEO) database. Differential expression analysis and weighted gene co-expression network analysis (WGCNA) were conducted on samples from patients with AMI and control samples to identify modules significantly associated with AMI. GO and KEGG analyses were applied to investigate the potential functions of these hub genes. Lastly, the mendelian randomization (MR) method was applied to analyze the causal relationship between the hub gene TNF and AMI. RESULTS 285 differentially expressed genes (DEGs) were identified through WCGNA and were clustered into 6 modules. The yellow module appeared most relevant to AMI. Further exploration through GO and KEGG pathway enrichment showed that key hub genes in the yellow module were linked to positive regulation of cytokine production, cytokine receptor binding, NF-kappa B signaling pathway, IL-17 signaling pathway, and TNF signaling pathway. The top 10 genes identified through Cytoscape software analysis were IL1B, TNF, TLR4, TLR2, FCGR3B, MMP9, CXCL8, TLR8, ICAM1, and JUK. Utilizing inverse variance weighting (IVW) analysis, we discovered a significant association between TNF and AMI risk, with an OR of 0.946 (95% CI = 0.911-0.984, p = 0.005). CONCLUSIONS The result of this study indicated that TNF, TLR2, TLR4, IL1B and FCGR3B may be potential biodiagnostic markers for AMI. TNF can inhibit inflammatory and oxidative stress responses in AMI, exerting a protective role in the heart.
Collapse
Affiliation(s)
- Xiaohe Yang
- Department of Cardiology, Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan, China
| | - Yingtao Huang
- Department of Orthopedics, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Dadong Tang
- School of Clinical College of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liangming Yue
- Department of Cardiology, Guangyuan Hospital of Traditional Chinese Medicine, Guangyuan, China
| |
Collapse
|
9
|
Karpov OA, Stotland A, Raedschelders K, Chazarin B, Ai L, Murray CI, Van Eyk JE. Proteomics of the heart. Physiol Rev 2024; 104:931-982. [PMID: 38300522 PMCID: PMC11381016 DOI: 10.1152/physrev.00026.2023] [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: 07/03/2023] [Revised: 12/25/2023] [Accepted: 01/14/2024] [Indexed: 02/02/2024] Open
Abstract
Mass spectrometry-based proteomics is a sophisticated identification tool specializing in portraying protein dynamics at a molecular level. Proteomics provides biologists with a snapshot of context-dependent protein and proteoform expression, structural conformations, dynamic turnover, and protein-protein interactions. Cardiac proteomics can offer a broader and deeper understanding of the molecular mechanisms that underscore cardiovascular disease, and it is foundational to the development of future therapeutic interventions. This review encapsulates the evolution, current technologies, and future perspectives of proteomic-based mass spectrometry as it applies to the study of the heart. Key technological advancements have allowed researchers to study proteomes at a single-cell level and employ robot-assisted automation systems for enhanced sample preparation techniques, and the increase in fidelity of the mass spectrometers has allowed for the unambiguous identification of numerous dynamic posttranslational modifications. Animal models of cardiovascular disease, ranging from early animal experiments to current sophisticated models of heart failure with preserved ejection fraction, have provided the tools to study a challenging organ in the laboratory. Further technological development will pave the way for the implementation of proteomics even closer within the clinical setting, allowing not only scientists but also patients to benefit from an understanding of protein interplay as it relates to cardiac disease physiology.
Collapse
Affiliation(s)
- Oleg A Karpov
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Aleksandr Stotland
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Koen Raedschelders
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Blandine Chazarin
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Lizhuo Ai
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Christopher I Murray
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| | - Jennifer E Van Eyk
- Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California, United States
| |
Collapse
|
10
|
An C, Li Z, Chen Y, Huang S, Yang F, Hu Y, Xu T, Zhang C, Ge S. The cGAS-STING pathway in cardiovascular diseases: from basic research to clinical perspectives. Cell Biosci 2024; 14:58. [PMID: 38720328 PMCID: PMC11080250 DOI: 10.1186/s13578-024-01242-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
The cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) synthase-stimulator of interferon genes (cGAS-STING) signaling pathway, an important component of the innate immune system, is involved in the development of several diseases. Ectopic DNA-induced inflammatory responses are involved in several pathological processes. Repeated damage to tissues and metabolic organelles releases a large number of damage-associated molecular patterns (mitochondrial DNA, nuclear DNA, and exogenous DNA). The DNA fragments released into the cytoplasm are sensed by the sensor cGAS to initiate immune responses through the bridging protein STING. Many recent studies have revealed a regulatory role of the cGAS-STING signaling pathway in cardiovascular diseases (CVDs) such as myocardial infarction, heart failure, atherosclerosis, and aortic dissection/aneurysm. Furthermore, increasing evidence suggests that inhibiting the cGAS-STING signaling pathway can significantly inhibit myocardial hypertrophy and inflammatory cell infiltration. Therefore, this review is intended to identify risk factors for activating the cGAS-STING pathway to reduce risks and to simultaneously further elucidate the biological function of this pathway in the cardiovascular field, as well as its potential as a therapeutic target.
Collapse
Affiliation(s)
- Cheng An
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China
| | - Zhen Li
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yao Chen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China
| | - Shaojun Huang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China
| | - Fan Yang
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ying Hu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Chengxin Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China.
| | - Shenglin Ge
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Anhui Medical University, No.218 Jixi Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
11
|
Fledderus J, Brouwer L, Kuiper T, Harmsen MC, Krenning G. H3K27Me3 abundance increases fibrogenesis during endothelial-to-mesenchymal transition via the silencing of microRNA-29c. Front Cardiovasc Med 2024; 11:1373279. [PMID: 38774662 PMCID: PMC11106376 DOI: 10.3389/fcvm.2024.1373279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/23/2024] [Indexed: 05/24/2024] Open
Abstract
Objective Endothelial-to-mesenchymal transition (EndMT) is a transdifferentiation process in which endothelial cells (ECs) adopt a mesenchymal-like phenotype. Over the past few years, it became clear that EndMT can contribute to several cardiovascular pathologies. However, the molecular pathways underlying the development of EndMT remain incompletely understood. Since the epigenetic enzyme Enhancer of Zeste Homolog 2 (EZH2) and its concomitant mark H3K27Me3 have been shown to be elevated in many cardiovascular diseases that associate with EndMT, we hypothesized that H3K27Me3 is a determinant for the susceptibility of EndMT. Methods To study the association between H3K27Me3 and EndMT, a knockdown model of EZH2 in human endothelial cells (HUVEC) was utilized to reduce H3K27Me3 abundance, followed by induction of EndMT using TGFβ1. The expression of molecular markers of EndMT and fibrogenesis were analysed. Results In cultured HUVECs, a reduction of H3K27Me3 abundance facilitates EndMT but mitigates fibrogenesis as shown by a decreased expression of collagen I and III. In HUVEC, H3K27Me3 abundance directly affects the expression of miR29c, a collagen-targeting miRNA. Additionally, knockdown of miR-29c in HUVEC with low H3K27Me3 abundance partly restored the expression of collagen I and III. Expectedly, in rats with perivascular fibrosis an increased abundance of H3K27Me3 associated with a decreased expression of miR-29c. Conclusion our data shows that endothelial fibrogenesis underlies an epigenetic regulatory pathway and we demonstrate that a decreased abundance of H3K27Me3 in ECs blunts fibrogenesis in part in a miR-29c dependent manner. Therefore, a reduction of H3K27Me3 could serve as a novel therapeutical strategy to mitigate fibrogenesis and may prove to be beneficial in fibrogenic diseases including atherosclerosis, cardiac fibrosis, and PAH.
Collapse
Affiliation(s)
- Jolien Fledderus
- Laboratory for Cardiovascular Regenerative Medicine, Medical Biology Section, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Linda Brouwer
- Laboratory for Cardiovascular Regenerative Medicine, Medical Biology Section, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Timara Kuiper
- Laboratory for Cardiovascular Regenerative Medicine, Medical Biology Section, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Martin C. Harmsen
- Laboratory for Cardiovascular Regenerative Medicine, Medical Biology Section, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Medical Biology Section, Department Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Division Experimental Pharmacology, Department Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
12
|
Ibarrola J, Xiang RR, Sun Z, Lu Q, Hill MA, Jaffe IZ. Inhibition of the histone methyltransferase EZH2 induces vascular stiffness. Clin Sci (Lond) 2024; 138:251-268. [PMID: 38362910 DOI: 10.1042/cs20231478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 02/17/2024]
Abstract
Vascular stiffness increases with aging, obesity and hypertension and predicts cardiovascular risk. The levels of histone H3-lysine-27 methylation (H3K27me) and the histone methyltransferase EZH2 both decrease in aging vessels, driving vascular stiffness. The impact of EZH2 inhibitors on vascular stiffness is unknown. We tested the hypothesis that the EZH2 inhibitor GSK126, currently in development for cancer treatment, increases vascular stiffness and explored underlying molecular mechanisms. Young (3 month) and middle-aged (12 month) male mice were treated with GSK126 for 1-2 months and primary human aortic smooth muscle cells (HASMCs) from young male and female donors were treated with GSK126 for 24-48 h. Stiffness was measured in vivo by pulse wave velocity and in vitro by atomic force microscopy (AFM) and vascular structure was quantified histologically. Extracellular matrix proteins were studied by qRT-PCR, immunoblotting, zymography and chromatin immunoprecipitation. GSK126 treatment decreased H3K27 methylation (H3K27me) and increased acetylation (H3K27ac) in mouse vessels and in HASMCs. In GSK126-treated mice, aortic stiffness increased without changes in vascular fibrosis. EZH2 inhibition enhanced elastin fiber degradation and matrix metalloprotease-2 (MMP2) expression. In HASMCs, GSK126 treatment increased synthetic phenotype markers and intrinsic HASMCs stiffness by AFM with altered cytoskeletal structure and increased nuclear actin staining. GSK126 also increased MMP2 protein expression, activity and enrichment of H3K27ac at the MMP2 promoter in HASMCs. GSK126 causes vascular stiffening, inducing MMP2 activity, elastin degradation, and modulation of SMC phenotype and cytoskeletal stiffness. These findings suggest that EZH2 inhibitors used to treat cancer could negatively impact the vasculature by enhancing stiffness and merits examination in human trials.
Collapse
Affiliation(s)
- Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, U.S.A
| | - Rachel R Xiang
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, U.S.A
| | - Zhe Sun
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65203, U.S.A
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, U.S.A
| | - Michael A Hill
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65203, U.S.A
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111, U.S.A
| |
Collapse
|
13
|
Wang T, Cheng M, Jin J, Bai Y, Zhang D, Zhang S, Xu J. Hypomethylation of the LncRNA H19 promoter accelerates osteogenic differentiation of vascular smooth muscle cells by activating the Erk1/2 pathways. J Int Med Res 2024; 52:3000605241234567. [PMID: 38530015 DOI: 10.1177/03000605241234567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024] Open
Abstract
OBJECTIVE Vascular calcification is a common chronic kidney disease complication. This study aimed to investigate the function of long non-coding RNA (LncRNA) H19 in vascular calcification to explore new therapeutic strategies. METHODS We induced osteogenic differentiation and calcification of vascular smooth muscle cells (VSMCs) using β-glycerophosphate. Then, we detected the LncRNA H19 promoter methylation status and Erk1/2 pathways using methylation-specific polymerase chain reaction and western blotting, respectively. RESULTS Compared with the control group, high phosphorus levels induced VSMC calcification, accompanied by increases in LncRNA H19 and the osteogenic marker Runx2 and reduction of the contractile phenotype marker SM22a. LncRNA H19 knockdown inhibited osteogenic differentiation and calcification of VSMCs. However, the suppressed role of VSMC calcification caused by shRNA H19 was partially reversed by simultaneous activation of the Erk1/2 pathways. Mechanically, we found that the methylation rate of CpG islands in the LncRNA H19 promoter region was significantly lower in the high-phosphorus group, and the hypomethylation state elevated LncRNA H19 levels, which in turn regulated phosphorylated Erk1/2 expression. CONCLUSIONS LncRNA H19 promoted osteogenic differentiation and calcification of VSMCs by regulating the Erk1/2 pathways. Additionally, hypomethylation of LncRNA H19 promoter CpG islands upregulated LncRNA H19 levels and subsequently activated Erk1/2 phosphorylation.
Collapse
Affiliation(s)
- Taoxia Wang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People's Republic of China
| | - Meijuan Cheng
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People's Republic of China
| | - Jingjing Jin
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People's Republic of China
| | - Yaling Bai
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People's Republic of China
| | - Dongxue Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People's Republic of China
| | - Shenglei Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People's Republic of China
| | - Jinsheng Xu
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, People's Republic of China
| |
Collapse
|
14
|
Olaniyi KS, Areloegbe SE, Fiemotongha FE. Cardiac energy depletion in a rat model of polycystic ovarian syndrome is reversed by acetate and associated with inhibitory effect of HDAC2/mTOR. Eur J Pharmacol 2024; 962:176243. [PMID: 38048978 DOI: 10.1016/j.ejphar.2023.176243] [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: 08/01/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/06/2023]
Abstract
In addition to the clinical manifestation of polycystic ovarian syndrome (PCOS), life-threatening diseases, especially hypertension and cardiovascular disease (CVD) are emerging critical complications of PCOS. Changes in cardiac energy remains an independent risk factor of CVD. Histone deacetylase (HDAC) inhibitors, including acetate has received attention for its beneficial role in energy regulation. Herein we hypothesized that acetate improves cardiac energy homeostasis in experimentally induced PCOS. Female Wistar rats (8-week-old) were divided into groups. To induce PCOS, 1 mg/kg of letrozole was given for 21 days. After confirmation of PCOS, acetate (200 mg/kg) was administered for 6 weeks. Rats with PCOS showed multiple ovarian cysts with androgen excess and decreased SHBG. The rats also manifested impaired glucose tolerance/hyperinsulinemia and hypertriglyceridemia. Increased systemic oxidative stress (malondialdehyde)/inflammatory (NF-kB/SDF-1) markers and nitric oxide deficiency (NO/eNOS) were observed. Though, the body weight was increased without affecting the cardiac mass index of PCOS rats. Nevertheless, there was an increase in cardiac triglyceride and oxidative stress/inflammatory markers with consequent cardiac injury, revealed by decreased levels of SIRT-1/HIF-1α and increased levels of CTGF/TGFβ-1 and plasma troponin T. These led to cardiac ATP depletion with increased AMP and AMP/ATP ratio. These alterations were accompanied by elevated levels of mTOR and HDAC2, which were reversed when treated with acetate. The present results interestingly suggest that HDAC2 inhibition by acetate reversed cardiac energy depletion and attendant cardiomorbidities in experimental PCOS model. A beneficial effect that is accompanied by suppressed expression of mTOR.
Collapse
Affiliation(s)
- Kehinde S Olaniyi
- Cardio/Endo-metabolic and Microbiome Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, 360101, Nigeria.
| | - Stephanie E Areloegbe
- Cardio/Endo-metabolic and Microbiome Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, 360101, Nigeria
| | - Faustina E Fiemotongha
- Cardio/Endo-metabolic and Microbiome Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, 360101, Nigeria
| |
Collapse
|
15
|
Geiger M, Gorica E, Mohammed SA, Mongelli A, Mengozi A, Delfine V, Ruschitzka F, Costantino S, Paneni F. Epigenetic Network in Immunometabolic Disease. Adv Biol (Weinh) 2024; 8:e2300211. [PMID: 37794610 DOI: 10.1002/adbi.202300211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/08/2023] [Indexed: 10/06/2023]
Abstract
Although a large amount of data consistently shows that genes affect immunometabolic characteristics and outcomes, epigenetic mechanisms are also heavily implicated. Epigenetic changes, including DNA methylation, histone modification, and noncoding RNA, determine gene activity by altering the accessibility of chromatin to transcription factors. Various factors influence these alterations, including genetics, lifestyle, and environmental cues. Moreover, acquired epigenetic signals can be transmitted across generations, thus contributing to early disease traits in the offspring. A closer investigation is critical in this aspect as it can help to understand the underlying molecular mechanisms further and gain insights into potential therapeutic targets for preventing and treating diseases arising from immuno-metabolic dysregulation. In this review, the role of chromatin alterations in the transcriptional modulation of genes involved in insulin resistance, systemic inflammation, macrophage polarization, endothelial dysfunction, metabolic cardiomyopathy, and nonalcoholic fatty liver disease (NAFLD), is discussed. An overview of emerging chromatin-modifying drugs and the importance of the individual epigenetic profile for personalized therapeutic approaches in patients with immuno-metabolic disorders is also presented.
Collapse
Affiliation(s)
- Martin Geiger
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Shafeeq Ahmed Mohammed
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessia Mongelli
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Alessandro Mengozi
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Valentina Delfine
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, University Hospital Zürich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- University Heart Center, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
- Department of Research and Education, University Hospital Zurich and University of Zürich, Wagistrasse 12, Schlieren, Zurich, 8952, Switzerland
| |
Collapse
|
16
|
Safdar M, Ullah M, Wahab A, Hamayun S, Ur Rehman M, Khan MA, Khan SU, Ullah A, Din FU, Awan UA, Naeem M. Genomic insights into heart health: Exploring the genetic basis of cardiovascular disease. Curr Probl Cardiol 2024; 49:102182. [PMID: 37913933 DOI: 10.1016/j.cpcardiol.2023.102182] [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: 10/23/2023] [Accepted: 10/28/2023] [Indexed: 11/03/2023]
Abstract
Cardiovascular diseases (CVDs) are considered as the leading cause of death worldwide. CVD continues to be a major cause of death and morbidity despite significant improvements in its detection and treatment. Therefore, it is strategically important to be able to precisely characterize an individual's sensitivity to certain illnesses. The discovery of genes linked to cardiovascular illnesses has benefited from linkage analysis and genome-wide association research. The last 20 years have seen significant advancements in the field of molecular genetics, particularly with the development of new tools like genome-wide association studies. In this article we explore the profound impact of genetic variations on disease development, prognosis, and therapeutic responses. And the significance of genetics in cardiovascular risk assessment and the ever-evolving realm of genetic testing, offering insights into the potential for personalized medicine in this domain. Embracing the future of cardiovascular care, the article explores the implications of pharmacogenomics for tailored treatments, the promise of emerging technologies in cardiovascular genetics and therapies, including the transformative influence of nanotechnology. Furthermore, it delves into the exciting frontiers of gene editing, such as CRISPR/Cas9, as a novel approach to combat cardiovascular diseases. And also explore the potential of stem cell therapy and regenerative medicine, providing a holistic view of the dynamic landscape of cardiovascular genomics and its transformative potential for the field of cardiovascular medicine.
Collapse
Affiliation(s)
- Mishal Safdar
- Department of Biological Sciences, National University of Medical Sciences (NUMS) Rawalpindi 46000, Punjab, Pakistan
| | - Muneeb Ullah
- Department of Pharmacy, Kohat University of Science, and technology (KUST), Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Abdul Wahab
- Department of Pharmacy, Kohat University of Science, and technology (KUST), Kohat, 26000, Khyber Pakhtunkhwa, Pakistan
| | - Shah Hamayun
- Department of Cardiology, Pakistan Institute of Medical Sciences (PIMS), Islamabad, 04485 Punjab, Pakistan
| | - Mahboob Ur Rehman
- Department of Cardiology, Pakistan Institute of Medical Sciences (PIMS), Islamabad, 04485 Punjab, Pakistan
| | - Muhammad Amir Khan
- Department of Foreign Medical education, Fergana Medical institute of Public Health, 2A Yangi Turon street, Fergana 150100, Uzbekistan
| | - Shahid Ullah Khan
- Department of Biochemistry, Women Medical and Dental College, Khyber Medical University, Abbottabad, 22080, Khyber Pakhtunkhwa, Pakistan
| | - Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Busan 48513, Republic of Korea
| | - Fakhar Ud Din
- Department of Pharmacy, Quaid-i-Azam University, 45320, Islamabad, Pakistan
| | - Uzma Azeem Awan
- Department of Biological Sciences, National University of Medical Sciences (NUMS) Rawalpindi 46000, Punjab, Pakistan
| | - Muhammad Naeem
- Department of Biological Sciences, National University of Medical Sciences (NUMS) Rawalpindi 46000, Punjab, Pakistan.
| |
Collapse
|
17
|
Zhang L, Xia C, Yang Y, Sun F, Zhang Y, Wang H, Liu R, Yuan M. DNA methylation and histone post-translational modifications in atherosclerosis and a novel perspective for epigenetic therapy. Cell Commun Signal 2023; 21:344. [PMID: 38031118 PMCID: PMC10688481 DOI: 10.1186/s12964-023-01298-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/27/2023] [Indexed: 12/01/2023] Open
Abstract
Atherosclerosis, which is a vascular pathology characterized by inflammation and plaque build-up within arterial vessel walls, acts as the important cause of most cardiovascular diseases. Except for a lipid-depository and chronic inflammatory, increasing evidences propose that epigenetic modifications are increasingly associated with atherosclerosis and are of interest from both therapeutic and biomarker perspectives. The chronic progressive nature of atherosclerosis has highlighted atherosclerosis heterogeneity and the fact that specific cell types in the complex milieu of the plaque are, by far, not the only initiators and drivers of atherosclerosis. Instead, the ubiquitous effects of cell type are tightly controlled and directed by the epigenetic signature, which, in turn, is affected by many proatherogenic stimuli, including low-density lipoprotein, proinflammatory, and physical forces of blood circulation. In this review, we summarize the role of DNA methylation and histone post-translational modifications in atherosclerosis. The future research directions and potential therapy for the management of atherosclerosis are also discussed. Video Abstract.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Chenhai Xia
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Yongjun Yang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Fangfang Sun
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Yu Zhang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Huan Wang
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China
| | - Rui Liu
- Department of Rehabilitation, Tangdu Hospital, Air Force Military Medical University, No. 1 Xinsi Road, Xi'an 710000, China.
| | - Ming Yuan
- Department of Cardiology, Xijing Hospital, Air Force Military Medical University, No. 127 Changle West Road, Xi'an, 710032, China.
| |
Collapse
|
18
|
Wu Z, Yu X, Zhang S, He Y, Guo W. Novel roles of PIWI proteins and PIWI-interacting RNAs in human health and diseases. Cell Commun Signal 2023; 21:343. [PMID: 38031146 PMCID: PMC10685540 DOI: 10.1186/s12964-023-01368-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Non-coding RNA has aroused great research interest recently, they play a wide range of biological functions, such as regulating cell cycle, cell proliferation, and intracellular substance metabolism. Piwi-interacting RNAs (piRNAs) are emerging small non-coding RNAs that are 24-31 nucleotides in length. Previous studies on piRNAs were mainly limited to evaluating the binding to the PIWI protein family to play the biological role. However, recent studies have shed more lights on piRNA functions; aberrant piRNAs play unique roles in many human diseases, including diverse lethal cancers. Therefore, understanding the mechanism of piRNAs expression and the specific functional roles of piRNAs in human diseases is crucial for developing its clinical applications. Presently, research on piRNAs mainly focuses on their cancer-specific functions but lacks investigation of their expressions and epigenetic modifications. This review discusses piRNA's biogenesis and functional roles and the recent progress of functions of piRNA/PIWI protein complexes in human diseases. Video Abstract.
Collapse
Affiliation(s)
- Zeyu Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Xiao Yu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China
| | - Yuting He
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China.
| | - Wenzhi Guo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Key Laboratory of Hepatobiliary and Pancreatic Surgery and Digestive Organ Transplantation of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Open and Key Laboratory of Hepatobiliary & Pancreatic Surgery and Digestive Organ Transplantation at Henan Universities, Zhengzhou, 450052, China.
- Henan Key Laboratory of Digestive Organ Transplantation, Zhengzhou, 450052, China.
| |
Collapse
|
19
|
Qadir Nanakali NM, Maleki Dana P, Sadoughi F, Asemi Z, Sharifi M, Asemi R, Yousefi B. The role of dietary polyphenols in alternating DNA methylation in cancer. Crit Rev Food Sci Nutr 2023; 63:12256-12269. [PMID: 35848113 DOI: 10.1080/10408398.2022.2100313] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Natural products such as curcumin, quercetin, and resveratrol have been shown to have antitumor effectsand several studies have examined their role in treating cancer, either alone or in combination with other chemotherapeutic drugs. These compounds are capable of affecting different cancer-related mechanisms, such as proliferation, inflammation, invasion, and metastasis. Along with all of the benefits of these agents, affecting epigenetic processes is one of the most important aspects of their impact. Epigenetic modifications can be categorized into three main processes that include DNA methylation, histone modification, and regulation of small non-coding RNAs. Therefore, targeting DNA methylation can be used as a cancer treatment strategy by identifying or developing methylation modulators. Herein, we take a look into the studies investigating the role of natural products (e.g. curcumin, resveratrol, epigallocatechin gallate (EGCG), and quercetin) in alternating the DNA methylation status of various cancer cells. We discuss how these compounds reduce the expression of enzymes mediating the methylation of tumor suppressor genes and thereby, increasing the expression of tumor suppressors while reactivating antitumor signaling pathways.
Collapse
Affiliation(s)
- Nadir Mustafa Qadir Nanakali
- Department of Biomedical Science, College of Science, Cihan University-Erbil, Kurdistan Region, Erbil, Iraq
- Department of Biology, College of Education, Salahaddin University-Erbil, Kurdistan Region, Erbil, Iraq
| | - Parisa Maleki Dana
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Fatemeh Sadoughi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, I.R. Iran
| | - Mehran Sharifi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Asemi
- Department of Internal Medicine, School of Medicine, Cancer Prevention Research Center, Seyyed Al-Shohada Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Bahman Yousefi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
20
|
Emon IM, Al-Qazazi R, Rauh MJ, Archer SL. The Role of Clonal Hematopoiesis of Indeterminant Potential and DNA (Cytosine-5)-Methyltransferase Dysregulation in Pulmonary Arterial Hypertension and Other Cardiovascular Diseases. Cells 2023; 12:2528. [PMID: 37947606 PMCID: PMC10650407 DOI: 10.3390/cells12212528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/12/2023] Open
Abstract
DNA methylation is an epigenetic mechanism that regulates gene expression without altering gene sequences in health and disease. DNA methyltransferases (DNMTs) are enzymes responsible for DNA methylation, and their dysregulation is both a pathogenic mechanism of disease and a therapeutic target. DNMTs change gene expression by methylating CpG islands within exonic and intergenic DNA regions, which typically reduces gene transcription. Initially, mutations in the DNMT genes and pathologic DNMT protein expression were found to cause hematologic diseases, like myeloproliferative disease and acute myeloid leukemia, but recently they have been shown to promote cardiovascular diseases, including coronary artery disease and pulmonary hypertension. We reviewed the regulation and functions of DNMTs, with an emphasis on somatic mutations in DNMT3A, a common cause of clonal hematopoiesis of indeterminant potential (CHIP) that may also be involved in the development of pulmonary arterial hypertension (PAH). Accumulation of somatic mutations in DNMT3A and other CHIP genes in hematopoietic cells and cardiovascular tissues creates an inflammatory environment that promotes cardiopulmonary diseases, even in the absence of hematologic disease. This review summarized the current understanding of the roles of DNMTs in maintenance and de novo methylation that contribute to the pathogenesis of cardiovascular diseases, including PAH.
Collapse
Affiliation(s)
- Isaac M. Emon
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (I.M.E.); (R.A.-Q.)
| | - Ruaa Al-Qazazi
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (I.M.E.); (R.A.-Q.)
| | - Michael J. Rauh
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Stephen L. Archer
- Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada; (I.M.E.); (R.A.-Q.)
| |
Collapse
|
21
|
Xu X, Sun B, Zhao C. Poly (ADP-Ribose) polymerase 1 and parthanatos in neurological diseases: From pathogenesis to therapeutic opportunities. Neurobiol Dis 2023; 187:106314. [PMID: 37783233 DOI: 10.1016/j.nbd.2023.106314] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023] Open
Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) is the most extensively studied member of the PARP superfamily, with its primary function being the facilitation of DNA damage repair processes. Parthanatos is a type of regulated cell death cascade initiated by PARP-1 hyperactivation, which involves multiple subroutines, including the accumulation of ADP-ribose polymers (PAR), binding of PAR and apoptosis-inducing factor (AIF), release of AIF from the mitochondria, the translocation of the AIF/macrophage migration inhibitory factor (MIF) complex, and massive MIF-mediated DNA fragmentation. Over the past few decades, the role of PARP-1 in central nervous system health and disease has received increasing attention. In this review, we discuss the biological functions of PARP-1 in neural cell proliferation and differentiation, memory formation, brain ageing, and epigenetic regulation. We then elaborate on the involvement of PARP-1 and PARP-1-dependant parthanatos in various neuropathological processes, such as oxidative stress, neuroinflammation, mitochondrial dysfunction, excitotoxicity, autophagy damage, and endoplasmic reticulum (ER) stress. Additional highlight contains PARP-1's implications in the initiation, progression, and therapeutic opportunities for different neurological illnesses, including neurodegenerative diseases, stroke, autism spectrum disorder (ASD), multiple sclerosis (MS), epilepsy, and neuropathic pain (NP). Finally, emerging insights into the repurposing of PARP inhibitors for the management of neurological diseases are provided. This review aims to summarize the exciting advancements in the critical role of PARP-1 in neurological disorders, which may open new avenues for therapeutic options targeting PARP-1 or parthanatos.
Collapse
Affiliation(s)
- Xiaoxue Xu
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China; Key Laboratory of Neurological Disease Big Data of Liaoning Province, Shenyang, China.
| | - Bowen Sun
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China; Key Laboratory of Neurological Disease Big Data of Liaoning Province, Shenyang, China
| | - Chuansheng Zhao
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China; Key Laboratory of Neurological Disease Big Data of Liaoning Province, Shenyang, China.
| |
Collapse
|
22
|
Tokarek J, Budny E, Saar M, Stańczak K, Wojtanowska E, Młynarska E, Rysz J, Franczyk B. Molecular Processes Involved in the Shared Pathways between Cardiovascular Diseases and Diabetes. Biomedicines 2023; 11:2611. [PMID: 37892985 PMCID: PMC10604380 DOI: 10.3390/biomedicines11102611] [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: 08/23/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 10/29/2023] Open
Abstract
Cardiovascular diseases and diabetes mellitus are currently among the diseases with the highest morbidity and mortality. The pathogenesis and development of these diseases remain strongly connected, along with inflammation playing a major role. Therefore, the treatment possibilities showing a positive impact on both of these diseases could be especially beneficial for patients. SGLT-2 inhibitors and GLP-1 receptor agonists present this dual effect. Moreover, the hostile composition of the gut microbiota could influence the progression of these conditions. In this review, the authors present the latest knowledge on and innovations in diabetes mellitus and CVD-with the focus on the molecular mechanisms and the role of the microbiota.
Collapse
Affiliation(s)
- Julita Tokarek
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (K.S.); (E.W.)
| | - Emilian Budny
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (K.S.); (E.W.)
| | - Maciej Saar
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (K.S.); (E.W.)
| | - Kamila Stańczak
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (K.S.); (E.W.)
| | - Ewa Wojtanowska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (K.S.); (E.W.)
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (K.S.); (E.W.)
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland (K.S.); (E.W.)
| |
Collapse
|
23
|
Zhen X, Zhao W, Wang J, Li L, He Y, Zhang J, Li C, Zhang S, Huang J, Luo B, Gao Y. Genetic Variations Within METTL16 and Susceptibility to Sudden Cardiac Death in Chinese Populations With Coronary Artery Disease. Am J Cardiol 2023; 202:90-99. [PMID: 37423176 DOI: 10.1016/j.amjcard.2023.06.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 07/11/2023]
Abstract
Despite recent advances in the prevention of coronary heart disease, the mortality rate of sudden cardiac death (SCD) remains high, which has become a substantial public health issue. Methyltransferase-like protein 16 (METTL16), as a newly discovered m6A methyltransferase, may be related to cardiovascular diseases. In the present study, a 6-base-pair insertion/deletion (del) polymorphism (rs58928048) in the METTL16 3'untranslated region (3'UTR) region was chosen as a candidate variant based on the findings of systematic screening. Then, the association between rs58928048 and susceptibility to SCD originating from coronary artery disease (SCD-CAD) in the Chinese population was investigated by conducting a case-control study that included 210 SCD-CAD cases and 644 matched healthy controls. Logistic regression analysis showed that the del allele of rs58928048 significantly reduced the SCD risk (odds ratio 0.69, 95% confidence interval 0.55 to 0.87, p = 0.00177). Genotype-phenotype correlation studies in human cardiac tissue samples demonstrated that the lower messenger RNA and protein expression levels of METTL16 were associated with the del allele of rs58928048. In the dual-luciferase activity assay, the del/del genotype exhibited lower transcriptional competence. Further bioinformatic analysis showed that the rs58928048 del variant may create transcription factor binding sites. Finally, pyrosequencing showed that the genotype of rs58928048 was related to the methylation status of the 3'UTR region of METTL16. Taken together, our findings provide evidence that rs58928048 may affect the methylation status of the 3'UTR region of METTL16 and subsequently affect its transcriptional activity thus as a potential genetic risk marker for SCD-CAD.
Collapse
Affiliation(s)
- Xiaoyuan Zhen
- Departments of Forensic Medicine, Medical College of Soochow University, Suzhou, China
| | - Wenfeng Zhao
- Departments of Forensic Medicine, Medical College of Soochow University, Suzhou, China
| | - Jiawen Wang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Lijuan Li
- Departments of Forensic Medicine, Medical College of Soochow University, Suzhou, China
| | - Yan He
- Departments of Epidemiology, Medical College of Soochow University, Suzhou, China
| | - Jianhua Zhang
- Department of Forensic Medicine, Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Sciences, Ministry of Justice, Shanghai, China
| | - Chengtao Li
- Department of Forensic Medicine, Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Sciences, Ministry of Justice, Shanghai, China
| | - Suhua Zhang
- Department of Forensic Medicine, Shanghai Key Laboratory of Forensic Medicine, Institute of Forensic Sciences, Ministry of Justice, Shanghai, China
| | - Jiang Huang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, China.
| | - Bin Luo
- Department of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Yuzhen Gao
- Departments of Forensic Medicine, Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
24
|
Martino F, Bassareo PP, Martino E, Romeo F, Calcaterra G, Perrone Filardi P, Indolfi C, Nodari S, Montemurro V, Guccione P, Salvo GD, Chessa M, Pedrinelli R, Mercuro G, Barillà F. Cardiovascular prevention in childhood: a consensus document of the Italian Society of Cardiology Working Group on Congenital Heart Disease and Cardiovascular Prevention in Paediatric Age. J Cardiovasc Med (Hagerstown) 2023; 24:492-505. [PMID: 37409595 DOI: 10.2459/jcm.0000000000001488] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Cardiovascular diseases (CVD) may be manifested from a very early age. Genetic and environmental (epigenetic) factors interact to affect development and give rise to an abnormal phenotypical expression of genetic information, although not eliciting changes in the nucleotide sequence of DNA. It has been scientifically proven that increased oxidative stress (OS) caused by disease (overweight, obesity, diabetes), nutritional imbalances, unhealthy lifestyles (smoking, alcohol, substance abuse) in the mother during pregnancy may induce placental dysfunction, intrauterine growth restriction, prematurity, low birth weight, postnatal adiposity rebound, metabolic alterations and consequent onset of traditional cardiovascular risk factors. OS represents the cornerstone in the onset of atherosclerosis and manifestation of CVD following an extended asymptomatic period. OS activates platelets and monocytes eliciting the release of pro-inflammatory, pro-atherogenic and pro-oxidising substances resulting in endothelial dysfunction, decrease in flow-mediated arterial dilatation and increase in carotid intima-media thickness. The prevention of CVD is defined as primordial (aimed at preventing risk factors development), primary (aimed at early identification and treatment of risk factors), secondary (aimed at reducing risk of future events in patients who have already manifested a cardiovascular event), and tertiary (aimed at limiting the complex outcome of disease). Atherosclerosis prevention should be implemented as early as possible. Appropriate screening should be carried out to identify children at high risk who are apparently healthy and implement measures including dietary and lifestyle changes, addition of nutritional supplements and, lastly, pharmacological treatment if risk profiles fail to normalise. Reinstating endothelial function during the reversible stage of atherosclerosis is crucial.
Collapse
Affiliation(s)
- Francesco Martino
- Department of Internal Clinical, Anesthesiological and Cardiovascular Sciences, La Sapienza University, Rome, Italy
| | - Pier Paolo Bassareo
- University College of Dublin, School of Medicine, Mater Misericordiae University Hospital and Children's Health Ireland at Crumlin, Dublin, Ireland
| | - Eliana Martino
- Department of Internal Clinical, Anesthesiological and Cardiovascular Sciences, La Sapienza University, Rome, Italy
| | | | | | | | - Ciro Indolfi
- Division of Cardiology, Research Centre for Cardiovascular Diseases, Magna Graecia University, Catanzaro
| | - Savina Nodari
- Department of Medical Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, ASST Spedali Civili, Brescia
| | | | - Paolo Guccione
- Department of Cardiology, Cardiac Surgery, Cardio-pulmonary Transplantation, IRCCS Bambino Gesu'Paediatric Hospital, Rome
| | - Giovanni Di Salvo
- Division of Paediatric Cardiology, Department of Women's and Children's Health, University of Padua, Padua
| | - Massimo Chessa
- ACHD UNIT, Pediatric and Adult Congenital Heart Centre, IRCCS-Policlinico San Donato, San Donato Milanese, Vita Salute San Raffaele University, Milan
| | - Roberto Pedrinelli
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa
| | | | | |
Collapse
|
25
|
Mongelli A, Mengozzi A, Geiger M, Gorica E, Mohammed SA, Paneni F, Ruschitzka F, Costantino S. Mitochondrial epigenetics in aging and cardiovascular diseases. Front Cardiovasc Med 2023; 10:1204483. [PMID: 37522089 PMCID: PMC10382027 DOI: 10.3389/fcvm.2023.1204483] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023] Open
Abstract
Mitochondria are cellular organelles which generate adenosine triphosphate (ATP) molecules for the maintenance of cellular energy through the oxidative phosphorylation. They also regulate a variety of cellular processes including apoptosis and metabolism. Of interest, the inner part of mitochondria-the mitochondrial matrix-contains a circular molecule of DNA (mtDNA) characterised by its own transcriptional machinery. As with genomic DNA, mtDNA may also undergo nucleotide mutations that have been shown to be responsible for mitochondrial dysfunction. During physiological aging, the mitochondrial membrane potential declines and associates with enhanced mitophagy to avoid the accumulation of damaged organelles. Moreover, if the dysfunctional mitochondria are not properly cleared, this could lead to cellular dysfunction and subsequent development of several comorbidities such as cardiovascular diseases (CVDs), diabetes, respiratory and cardiovascular diseases as well as inflammatory disorders and psychiatric diseases. As reported for genomic DNA, mtDNA is also amenable to chemical modifications, namely DNA methylation. Changes in mtDNA methylation have shown to be associated with altered transcriptional programs and mitochondrial dysfunction during aging. In addition, other epigenetic signals have been observed in mitochondria, in particular the interaction between mtDNA methylation and non-coding RNAs. Mitoepigenetic modifications are also involved in the pathogenesis of CVDs where oxygen chain disruption, mitochondrial fission, and ROS formation alter cardiac energy metabolism leading to hypertrophy, hypertension, heart failure and ischemia/reperfusion injury. In the present review, we summarize current evidence on the growing importance of epigenetic changes as modulator of mitochondrial function in aging. A better understanding of the mitochondrial epigenetic landscape may pave the way for personalized therapies to prevent age-related diseases.
Collapse
Affiliation(s)
- Alessia Mongelli
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zürich, Zurich, Switzerland
| | - Alessandro Mengozzi
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zürich, Zurich, Switzerland
| | - Martin Geiger
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zürich, Zurich, Switzerland
| | - Era Gorica
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zürich, Zurich, Switzerland
| | - Shafeeq Ahmed Mohammed
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zürich, Zurich, Switzerland
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zürich, Zurich, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Frank Ruschitzka
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zürich, Zurich, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, Zurich University Hospital and University of Zürich, Zurich, Switzerland
- Department of Cardiology, University Heart Center, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Spezzini J, Piragine E, d'Emmanuele di Villa Bianca R, Bucci M, Martelli A, Calderone V. Hydrogen sulfide and epigenetics: Novel insights into the cardiovascular effects of this gasotransmitter. Br J Pharmacol 2023; 180:1793-1802. [PMID: 37005728 DOI: 10.1111/bph.16083] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/20/2023] [Accepted: 03/28/2023] [Indexed: 04/04/2023] Open
Abstract
Epigenetics studies the heritable modifications of genome expression that do not affect the nucleotide sequence. Epigenetic modifications can be divided into: DNA methylation, histone modifications, and modulation of genome expression by non-coding RNAs. Alteration of these mechanisms can alter the phenotype, and can lead to disease onset. The endogenous gasotransmitter hydrogen sulfide (H2 S) plays pleiotropic roles in many systems, including the cardiovascular (CV) system, and its mechanism of action mainly includes S-persulfidation of cysteine residues. Recent evidence suggests that many H2 S-mediated biological activities are based on the epigenetic regulation of cellular function, with effects ranging from DNA methylation to modification of histones and regulation of non-coding RNAs. This review describes the role of H2 S-regulating epigenetic mechanisms, providing a panorama of the current literature, and offers a novel scenario for the development of H2 S-releasing 'epidrugs' with a potential clinical use in the prevention and treatment of many CV and non-CV disorders.
Collapse
Affiliation(s)
| | | | | | - Mariarosaria Bucci
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, Pisa, Italy
| | | |
Collapse
|
27
|
Peruzzi M, Covi K, Saccucci M, Pingitore A, Saade W, Sciarra L, Cristalli MP, Miraldi F, Frati G, Cavarretta E. Current knowledge on the association between cardiovascular and periodontal disease: an umbrella review. Minerva Cardiol Angiol 2023; 71:208-220. [PMID: 35332749 DOI: 10.23736/s2724-5683.22.06022-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Periodontal disease (PD) and cardiovascular diseases (CVD) are among the most common pathologies in the world and their relationship has long been studied. Both conditions lead to a chronic inflammatory process with degenerative characteristics and their biunivocal correlation is now well established. The aim of this umbrella review on cardiovascular and periodontal disease was to evaluate the real degree of association between these two pathological conditions. EVIDENCE ACQUISITION We conducted a comprehensive literature search on PubMed/Medline and in the Cochrane Library for systematic reviews focused on clinical evidence regarding the relationship between PD and CVD. The internal validity of systematic reviews and meta-analyses was formally analyzed using the Overview Quality Assessment Questionnaire (OQAQ) Tool. The umbrella review was planned in accordance with current international recommendations and was described as specified by the PRISMA Guidelines. EVIDENCE SYNTHESIS Thirty-one systematic reviews, including 8 meta-analyses for a total of 507 clinical studies and over 3,549,966 patients were included. PD resulted to be associated with a higher risk of developing CVD (acute coronary syndrome, acute myocardial infarction) and cerebrovascular diseases (ischemic stroke); however, if the treatment of periodontitis reduces the risk of CVD events related is yet to be investigated. CONCLUSIONS To date, the relationship between CVD and PD provides heterogeneous data. There is an association between PD and CVD, but a causal relationship cannot be established. Further research with properly designed long-term follow-up studies are needed in order to examine various physiopathological aspects of their association.
Collapse
Affiliation(s)
- Mariangela Peruzzi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University, Rome, Italy
- Mediterranea Cardiocentro, Naples, Italy
| | - Karin Covi
- Department of Oral and Maxillo Facial Sciences, Sapienza University, Rome, Italy
| | - Matteo Saccucci
- Department of Oral and Maxillo Facial Sciences, Sapienza University, Rome, Italy
| | - Annachiara Pingitore
- Paride Stefanini Department of General and Specilized Surgery, Sapienza University, Rome, Italy
| | - Wael Saade
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Luigi Sciarra
- Department of Clinical Medicine, Public Health, Life and Environment Sciences, University of L'Aquila, L'Aquila, Italy
| | - Maria P Cristalli
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
| | - Fabio Miraldi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
- IRCCS Neuromed, Località Camerelle, Pozzilli, Isernia, Italy
| | - Elena Cavarretta
- Mediterranea Cardiocentro, Naples, Italy -
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
| |
Collapse
|
28
|
Song B, Xie B, Liu M, Li H, Shi D, Zhao F. Bibliometric and visual analysis of RAN methylation in cardiovascular disease. Front Cardiovasc Med 2023; 10:1110718. [PMID: 37063953 PMCID: PMC10098125 DOI: 10.3389/fcvm.2023.1110718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 03/07/2023] [Indexed: 03/31/2023] Open
Abstract
BackgroundRNA methylation is associated with cardiovascular disease (CVD) occurrence and development. The purpose of this study is to visually analyze the results and research trends of global RNA methylation in CVD.MethodsArticles and reviews on RNA methylation in CVD published before 6 November 2022 were searched in the Web of Science Core Collection. Visual and statistical analysis was performed using CiteSpace 1.6.R4 advanced and VOSviewer 1.6.18.ResultsThere were 847 papers from 1,188 institutions and 63 countries/regions. Over approximately 30 years, there was a gradual increase in publications and citations on RNA methylation in CVD. America and China had the highest output (284 and 259 papers, respectively). Nine of the top 20 institutions that published articles were from China, among which Fudan University represented the most. The International Journal of Molecular Sciences was the journal with the most studies. Nature was the most co-cited journal. The most influential writers were Zhang and Wang from China and Mathiyalagan from the United States. After 2015, the primary keywords were cardiac development, heart, promoter methylation, RNA methylation, and N6-methyladenosine. Nuclear RNA, m6A methylation, inhibition, and myocardial infarction were the most common burst keywords from 2020 to the present.ConclusionsA bibliometric analysis reveals research hotspots and trends of RNA methylation in CVD. The regulatory mechanisms of RNA methylation related to CVD and the clinical application of their results, especially m6A methylation, are likely to be the focus of future research.
Collapse
Affiliation(s)
- Boce Song
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Beili Xie
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Mingwang Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haohao Li
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Dazhuo Shi
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
| | - Fuhai Zhao
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- National Clinical Research Center for Chinese Medicine Cardiology, Beijing, China
- Correspondence: Fuhai Zhao
| |
Collapse
|
29
|
Kefayati F, Karimi Babaahmadi A, Mousavi T, Hodjat M, Abdollahi M. Epigenotoxicity: a danger to the future life. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART A, TOXIC/HAZARDOUS SUBSTANCES & ENVIRONMENTAL ENGINEERING 2023; 58:382-411. [PMID: 36942370 DOI: 10.1080/10934529.2023.2190713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/17/2023] [Accepted: 02/21/2023] [Indexed: 06/18/2023]
Abstract
Environmental toxicants can regulate gene expression in the absence of DNA mutations via epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNAs' (ncRNAs). Here, all three epigenetic modifications for seven important categories of diseases and the impact of eleven main environmental factors on epigenetic modifications were discussed. Epigenetic-related mechanisms are among the factors that could explain the root cause of a wide range of common diseases. Its overall impression on the development of diseases can help us diagnose and treat diseases, and besides, predict transgenerational and intergenerational effects. This comprehensive article attempted to address the relationship between environmental factors and epigenetic modifications that cause diseases in different categories. The studies main gap is that the precise role of environmentally-induced epigenetic alterations in the etiology of the disorders is unknown; thus, still more well-designed researches need to be accomplished to fill this gap. The present review aimed to first summarize the adverse effect of certain chemicals on the epigenome that may involve in the onset of particular disease based on in vitro and in vivo models. Subsequently, the possible adverse epigenetic changes that can lead to many human diseases were discussed.
Collapse
Affiliation(s)
- Farzaneh Kefayati
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Atoosa Karimi Babaahmadi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Taraneh Mousavi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahshid Hodjat
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Dental Research Center, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
30
|
Zhao M, Liu J, Xin M, Yang K, Huang H, Zhang W, Zhang J, He S. Pulmonary arterial hypertension associated with congenital heart disease: An omics study. Front Cardiovasc Med 2023; 10:1037357. [PMID: 36970344 PMCID: PMC10036813 DOI: 10.3389/fcvm.2023.1037357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
Pulmonary arterial hypertension associated with congenital heart disease (PAH-CHD) is a severely progressive condition with uncertain physiological course. Hence, it has become increasingly relevant to clarify the specific mechanisms of molecular modification, which is crucial to identify more treatment strategies. With the rapid development of high-throughput sequencing, omics technology gives access to massive experimental data and advanced techniques for systems biology, permitting comprehensive assessment of disease occurrence and progression. In recent years, significant progress has been made in the study of PAH-CHD and omics. To provide a comprehensive description and promote further in-depth investigation of PAH-CHD, this review attempts to summarize the latest developments in genomics, transcriptomics, epigenomics, proteomics, metabolomics, and multi-omics integration.
Collapse
Affiliation(s)
- Maolin Zhao
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Jian Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Mei Xin
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Ke Yang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Honghao Huang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Wenxin Zhang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Jinbao Zhang
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
| | - Siyi He
- Department of Cardiovascular Surgery, Affiliated Hospital of Southwest Jiaotong University, General Hospital of Western Theater Command, Chengdu, China
- Correspondence: Siyi He
| |
Collapse
|
31
|
Inorganic Pyrophosphate Plasma Levels Are Decreased in Pseudoxanthoma Elasticum Patients and Heterozygous Carriers but Do Not Correlate with the Genotype or Phenotype. J Clin Med 2023; 12:jcm12051893. [PMID: 36902680 PMCID: PMC10003929 DOI: 10.3390/jcm12051893] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 01/29/2023] [Accepted: 02/10/2023] [Indexed: 03/08/2023] Open
Abstract
Pseudoxanthoma elasticum (PXE) is a rare ectopic calcification disorder affecting soft connective tissues that is caused by biallelic ABCC6 mutations. While the underlying pathomechanisms are incompletely understood, reduced circulatory levels of inorganic pyrophosphate (PPi)-a potent mineralization inhibitor-have been reported in PXE patients and were suggested to be useful as a disease biomarker. In this study, we explored the relation between PPi, the ABCC6 genotype and the PXE phenotype. For this, we optimized and validated a PPi measurement protocol with internal calibration that can be used in a clinical setting. An analysis of 78 PXE patients, 69 heterozygous carriers and 14 control samples revealed significant differences in the measured PPi levels between all three cohorts, although there was overlap between all groups. PXE patients had a ±50% reduction in PPi levels compared to controls. Similarly, we found a ±28% reduction in carriers. PPi levels were found to correlate with age in PXE patients and carriers, independent of the ABCC6 genotype. No correlations were found between PPi levels and the Phenodex scores. Our results suggest that other factors besides PPi are at play in ectopic mineralization, which limits the use of PPi as a predictive biomarker for severity and disease progression.
Collapse
|
32
|
Škrlec I. Circadian system microRNAs - Role in the development of cardiovascular diseases. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 137:225-267. [PMID: 37709378 DOI: 10.1016/bs.apcsb.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Circadian rhythm regulates numerous physiological processes, and disruption of the circadian clock can lead to cardiovascular disease. Cardiovascular disease is the leading cause of morbidity and mortality worldwide. Small non-coding RNAs, microRNAs (miRNAs), are involved in regulating gene expression, both those important for the cardiovascular system and key circadian clock genes. Epigenetic mechanisms based on miRNAs are essential for fine-tuning circadian physiology. Indeed, some miRNAs depend on circadian periodicity, others are under the influence of light, and still others are under the influence of core clock genes. Dysregulation of miRNAs involved in circadian rhythm modulation has been associated with inflammatory conditions of the endothelium and atherosclerosis, which can lead to coronary heart disease and myocardial infarction. Epigenetic processes are reversible through their association with environmental factors, enabling innovative preventive and therapeutic strategies for cardiovascular disease. Here, is a review of recent findings on how miRNAs modulate circadian rhythm desynchronization in cardiovascular disease. In the era of personalized medicine, the possibility of treatment with miRNA antagomirs should be time-dependent to correspond to chronotherapy and achieve the most significant efficacy.
Collapse
Affiliation(s)
- Ivana Škrlec
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia.
| |
Collapse
|
33
|
The Nine RNA Methylation Regulatory Gene Signature Is Associated with the Pathogenesis of Atrial Fibrillation by Modulating the Immune Microenvironment in the Atrial Tissues. DISEASE MARKERS 2023. [DOI: 10.1155/2023/7277369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Background. Atrial fibrillation (AF) is the most common type of cardiac arrhythmias and a major cause of cardiovascular disease (CVD)-related deaths globally. RNA methylation is the most frequent posttranscriptional modification in the eukaryotic RNAs. Previous studies have demonstrated close associations between the status of RNA methylation and CVD. Methods. We comprehensively evaluated the relationship between RNA methylation and AF. Least absolute shrinkage and selection operator (LASSO) logistic regression analysis was used to establish a risk score model in AF. Biological functional analysis was used to explore the relationship between RNA methylation related signatures and immune microenvironment characteristics. Machine learning was used to recognize the outstanding RNA methylation regulators in AF. Results. There was a significant variant of the mRNA expression of RNA methylation regulators in AF. RNA methylation related risk score could predict the onset of AF and closely associated with immune microenvironment features. XG-Boost algorithm and SHAP recognized that NSUN3 and DCPS might play a key role in the development of AF. Meanwhile, NSUN3 and DCPS had potential diagnostic value in AF. Conclusion. RNA methylation regulatory genes are associated with the onset of AF by modulating the immune microenvironment. The nine AF risk-related RNA methylation regulatory gene signature is a potential diagnostic biomarker and therapeutic target for AF.
Collapse
|
34
|
Aljahdali AA, Goodrich JM, Dolinoy DC, Kim HM, Ruiz-Narváez EA, Baylin A, Cantoral A, Torres-Olascoaga LA, Téllez-Rojo MM, Peterson KE. DNA Methylation Is a Potential Biomarker for Cardiometabolic Health in Mexican Children and Adolescents. EPIGENOMES 2023; 7:4. [PMID: 36810558 PMCID: PMC9944859 DOI: 10.3390/epigenomes7010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/28/2023] [Accepted: 01/29/2023] [Indexed: 02/05/2023] Open
Abstract
DNA methylation (DNAm) is a plausible mechanism underlying cardiometabolic abnormalities, but evidence is limited among youth. This analysis included 410 offspring of the Early Life Exposure in Mexico to Environmental Toxicants (ELEMENT) birth cohort followed up to two time points in late childhood/adolescence. At Time 1, DNAm was quantified in blood leukocytes at long interspersed nuclear elements (LINE-1), H19, and 11β-hydroxysteroid dehydrogenase type 2 (11β-HSD-2), and at Time 2 in peroxisome proliferator-activated receptor alpha (PPAR-α). At each time point, cardiometabolic risk factors were assessed including lipid profiles, glucose, blood pressure, and anthropometry. Linear mixed effects models were used for LINE-1, H19, and 11β-HSD-2 to account for the repeated-measure outcomes. Linear regression models were conducted for the cross-sectional association between PPAR-α with the outcomes. DNAm at LINE-1 was associated with log glucose at site 1 [β = -0.029, p = 0.0006] and with log high-density lipoprotein cholesterol at site 3 [β = 0.063, p = 0.0072]. 11β-HSD-2 DNAm at site 4 was associated with log glucose (β = -0.018, p = 0.0018). DNAm at LINE-1 and 11β-HSD-2 was associated with few cardiometabolic risk factors among youth in a locus-specific manner. These findings underscore the potential for epigenetic biomarkers to increase our understanding of cardiometabolic risk earlier in life.
Collapse
Affiliation(s)
- Abeer A. Aljahdali
- Department of Clinical Nutrition, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jaclyn M. Goodrich
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| | - Dana C. Dolinoy
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| | - Hyungjin M. Kim
- Center for Computing, Analytics and Research, University of Michigan, Ann Arbor, MI 48109, USA
| | | | - Ana Baylin
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Epidemiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Alejandra Cantoral
- Department of Health, Iberoamericana University, Mexico City 01219, Mexico
| | - Libni A. Torres-Olascoaga
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca 62100, Mexico
| | - Martha M. Téllez-Rojo
- Center for Nutrition and Health Research, National Institute of Public Health, Cuernavaca 62100, Mexico
| | - Karen E. Peterson
- Department of Nutritional Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Environmental Health Sciences, University of Michigan, 1415 Washington Heights, Ann Arbor, MI 48109, USA
| |
Collapse
|
35
|
Donniacuo M, De Angelis A, Telesca M, Bellocchio G, Riemma MA, Paolisso P, Scisciola L, Cianflone E, Torella D, Castaldo G, Capuano A, Urbanek K, Berrino L, Rossi F, Cappetta D. Atrial fibrillation: Epigenetic aspects and role of sodium-glucose cotransporter 2 inhibitors. Pharmacol Res 2023; 188:106591. [PMID: 36502999 DOI: 10.1016/j.phrs.2022.106591] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/30/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022]
Abstract
Atrial fibrillation (AF) is the most frequent arrhythmia and is associated with substantial morbidity and mortality. Pathophysiological aspects consist in the activation of pro-fibrotic signaling and Ca2+ handling abnormalities at atrial level. Structural and electrical remodeling creates a substrate for AF by triggering conduction abnormalities and cardiac arrhythmias. The care of AF patients focuses predominantly on anticoagulation, symptoms control and the management of risk factors and comorbidities. The goal of AF therapy points to restore sinus rhythm, re-establish atrioventricular synchrony and improve atrial contribution to the stroke volume. New layer of information to better comprehend AF pathophysiology, and identify targets for novel pharmacological interventions consists of the epigenetic phenomena including, among others, DNA methylation, histone modifications and noncoding RNAs. Moreover, the benefits of sodium-glucose cotransporter 2 inhibitors (SGLT2i) in diabetic and non-diabetic patients at cardiovascular risk as well as emerging evidence on the ability of SGLT2i to modify epigenetic signature in cardiovascular diseases provide a solid background to investigate a possible role of this drug class in the onset and progression of AF. In this review, following a summary of pathophysiology and management, epigenetic mechanisms in AF and the potential of sodium-glucose SGLT2i in AF patients are discussed.
Collapse
Affiliation(s)
- M Donniacuo
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - A De Angelis
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - M Telesca
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - G Bellocchio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - M A Riemma
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - P Paolisso
- Cardiovascular Center Aalst, OLV Hospital, Aalst, Belgium; Department of Advanced Biomedical Sciences, University of Naples "Federico II", Via A. Pansini 5, 80131 Naples, Italy
| | - L Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - E Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - D Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Viale Europa, 88100 Catanzaro, Italy
| | - G Castaldo
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Via A. Pansini 5, 80131 Naples, Italy; CEINGE-Advanced, Via G. Salvatore 486, 80131 Naples, Italy
| | - A Capuano
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - K Urbanek
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Via A. Pansini 5, 80131 Naples, Italy; CEINGE-Advanced, Via G. Salvatore 486, 80131 Naples, Italy.
| | - L Berrino
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - F Rossi
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - D Cappetta
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
36
|
Wasiak S, Tsujikawa LM, Daze E, Gilham D, Stotz SC, Rakai BD, Sarsons CD, Fu L, Azhar S, Jahagirdar R, Sweeney M, Johansson JO, Wong NCW, Kulikowski E. Epigenetic BET reader inhibitor apabetalone (RVX-208) counters proinflammatory aortic gene expression in a diet induced obesity mouse model and in human endothelial cells. Atherosclerosis 2023; 364:10-19. [PMID: 36455344 DOI: 10.1016/j.atherosclerosis.2022.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 10/06/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND AND AIMS Obese patients are at risk for type 2 diabetes mellitus (T2DM) and cardiovascular disease (CVD). A lipid-rich diet promotes arterial changes by inducing hypertension, oxidative stress, and inflammation. Bromodomain and extraterminal (BET) proteins contribute to endothelial and immune cell activation in vitro and in atherosclerosis mouse models. We aim to determine if BET inhibition can reduce lipid-rich diet-induced vascular inflammation in mice. METHODS Body weight, serum glucose and lipid levels were measured in mice fed a high-fat diet (HFD) or low-fat diet (LFD) for 6 weeks and at study termination. BET inhibitors apabetalone and JQ1 were co-administered with the HFD for additional 16 weeks. Aortic gene expression was analyzed post necropsy by PCR, Nanostring nCounter® Inflammation Panel and bioinformatics pathway analysis. Transcription changes and BRD4 chromatin occupancy were analyzed in primary human endothelial cells in response to TNFα and apabetalone. RESULTS HFD induced weight gain, visceral obesity, high fasting blood glucose, glucose intolerance and insulin resistance compared to LFD controls. HFD upregulated the aortic expression of 47 genes involved in inflammation, innate immunity, cytoskeleton and complement pathways. Apabetalone and JQ1 treatment reduced HFD-induced aortic expression of proinflammatory genes. Congruently, bioinformatics predicted enhanced signaling by TNFα in the HFD versus LFD aorta, which was countered by BETi treatment. TNFα-stimulated human endothelial cells had increased expression of HFD-sensitive genes and higher BRD4 chromatin occupancy, which was countered by apabetalone treatment. CONCLUSIONS HFD induces vascular inflammation in mice through TNFα signaling. Apabetalone treatment reduces this proinflammatory phenotype, providing mechanistic insight into how BET inhibitors may reduce CVD risk in obese patients.
Collapse
Affiliation(s)
- Sylwia Wasiak
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Laura M Tsujikawa
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Emily Daze
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Dean Gilham
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Stephanie C Stotz
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Brooke D Rakai
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Chris D Sarsons
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Li Fu
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Salman Azhar
- VA Palo Alto Health Care System, 3801 Miranda Avenue, Palo Alto, CA, 94304, USA
| | - Ravi Jahagirdar
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Michael Sweeney
- Resverlogix Inc., 535 Mission St, 14th Floor, San Francisco, CA, 94105
| | - Jan O Johansson
- Resverlogix Inc., 535 Mission St, 14th Floor, San Francisco, CA, 94105
| | - Norman C W Wong
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Ewelina Kulikowski
- Resverlogix Corp., 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada.
| |
Collapse
|
37
|
Mansouri E, Esmaeili F, Montaseri M, Emami MA, Koochakkhani S, Khayatian M, Zarei H, Turki H, Eftekhar E. Association of methylation status of ABCA1/G1 genes with the risk of coronary artery disease. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00381-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Abstract
Background
ATP-binding cassette transporters A1/G1 (ABCA1/G1) is a main regulator of HDL (high-density lipoprotein) formation and reverse cholesterol transport. Impaired ABCA1/G1 genes function may seriously affect cholesterol homeostasis, leading to increased risk of cardiovascular disease. In the present study, the association of ABCA1/G1 genes methylation status with the risk of coronary artery disease (CAD), risk factors of CAD, and serum level of lipid parameters was investigated.
This study was conducted on 70 CAD patients and 40 control subjects. All CAD subjects with diabetes mellitus were excluded. The promoter methylation status of ABCA1/G1 genes was determined by the methylation-specific polymerase chain reaction (MS-PCR) method and serum lipid parameters were assessed using commercial kits.
Results
ABCA1 promoter methylation was higher in CAD group compared to the control participants (80% vs. 60%). Hypermethylation of the ABCA1 gene significantly increases the risk of CAD in the total population (OR 3.886, 95% CI (1.181–12.791), p = 0.026). ABCG1 methylation status showed no difference between CAD and control subjects. In addition, no significant association was noted between methylation status of ABCA1/G1 and serum level of lipid profile.
Conclusions
Altogether, our study shows that ABCA1 gene promoter hypermethylation may increase the risk of CAD, which may help identify people at risk of developing CAD.
Collapse
|
38
|
Yu Q, Yang J, Wang J, Yu R, Li J, Cheng J, Hu Y, Li Z, Zheng N, Zhang Z, Li X, Wang Y, Du W, Zhu K, Chen X, Su J. DNA methylation profile in the whole blood of acute coronary syndrome patients with aspirin resistance. J Clin Lab Anal 2022; 37:e24821. [PMID: 36550638 PMCID: PMC9833987 DOI: 10.1002/jcla.24821] [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: 11/16/2022] [Revised: 12/07/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Aspirin resistance (AR) results in major adverse cardiovascular events, and DNA methylation might participate in the regulation of this pathological process. METHODS In present study, a sum of 35 patients with AR and 35 non-AR (NAR) controls were enrolled. Samples from 5 AR and 5 NAR were evaluated in an 850 BeadChip DNA methylation assay, and another 30 AR versus 30 NAR were evaluated to validate the differentially methylated CpG loci (DML). Then, qRT-PCR was used to investigate the target mRNA expression of genes at CpG loci. Finally, Gene Ontology (GO) as well as Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to reveal the enriched pathways. RESULTS The AR and NAR groups displayed significant differences in DNA methylation at 7707 positions, with 270 hypermethylated sites (e.g., cg09555818 located in APOC2) and 7437 sites hypomethylated sites (e.g., cg26828689 located in SLC12A5). Six DML were validated by pyrosequencing, and it was confirmed that DNA methylation (cg16391727, cg21008208, cg21293749, and cg13945576) was related to the increasing risk of AR. The relative mRNA expression of the ROR1 gene was also associated with AR (p = 0.007), suggesting that the change of cg21293749 in DNA methylation might lead to differential ROR1 mRNA expression, ultimately resulting in AR. Furthermore, the identified differentially methylated sites were associated with the molecular pathways such as circadian rhythms and insulin secretion. CONCLUSION Hence, the distinct DNA methylation might play a vital role in the biological regulation of AR through the pathways such as circadian rhythms.
Collapse
Affiliation(s)
- Qinglin Yu
- Department of Traditional Chinese Internal MedicineNingbo No. 1 HospitalNingboChina
| | - Jin Yang
- Department of GeriatricsNingbo No. 1 HospitalNingboChina
| | - Jiang Wang
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Ruoyan Yu
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Jiyi Li
- Department of CardiologyYuyao People's Hospital of Zhejiang ProvinceYuyaoChina
| | - Ji Cheng
- Department of Emergency, HwaMei HospitalUniversity of Chinese Academy of SciencesNingboChina
| | - Yingchu Hu
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Zhenwei Li
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Nan Zheng
- Department of Cardiology, HwaMei HospitalUniversity of Chinese Academy of SciencesNingboChina
| | - Zhaoxia Zhang
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Xiaojing Li
- Department of GeriatricsNingbo No. 1 HospitalNingboChina
| | - Yong Wang
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Weiping Du
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Keqi Zhu
- Department of Traditional Chinese Internal MedicineNingbo No. 1 HospitalNingboChina
| | - Xiaomin Chen
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Jia Su
- Department of CardiologyNingbo No.1 HospitalNingboChina,Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| |
Collapse
|
39
|
Fadaei S, Zarepour F, Parvaresh M, Motamedzadeh A, Tamehri Zadeh SS, Sheida A, Shabani M, Hamblin MR, Rezaee M, Zarei M, Mirzaei H. Epigenetic regulation in myocardial infarction: Non-coding RNAs and exosomal non-coding RNAs. Front Cardiovasc Med 2022; 9:1014961. [PMID: 36440025 PMCID: PMC9685618 DOI: 10.3389/fcvm.2022.1014961] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/17/2022] [Indexed: 08/13/2023] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of deaths globally. The early diagnosis of MI lowers the rate of subsequent complications and maximizes the benefits of cardiovascular interventions. Many efforts have been made to explore new therapeutic targets for MI, and the therapeutic potential of non-coding RNAs (ncRNAs) is one good example. NcRNAs are a group of RNAs with many different subgroups, but they are not translated into proteins. MicroRNAs (miRNAs) are the most studied type of ncRNAs, and have been found to regulate several pathological processes in MI, including cardiomyocyte inflammation, apoptosis, angiogenesis, and fibrosis. These processes can also be modulated by circular RNAs and long ncRNAs via different mechanisms. However, the regulatory role of ncRNAs and their underlying mechanisms in MI are underexplored. Exosomes play a crucial role in communication between cells, and can affect both homeostasis and disease conditions. Exosomal ncRNAs have been shown to affect many biological functions. Tissue-specific changes in exosomal ncRNAs contribute to aging, tissue dysfunction, and human diseases. Here we provide a comprehensive review of recent findings on epigenetic changes in cardiovascular diseases as well as the role of ncRNAs and exosomal ncRNAs in MI, focusing on their function, diagnostic and prognostic significance.
Collapse
Affiliation(s)
- Sara Fadaei
- Department of Internal Medicine and Endocrinology, Shohadae Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Zarepour
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mehrnoosh Parvaresh
- Department of Physical Medicine and Rehabilitation, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Alireza Motamedzadeh
- Department of Internal Medicine, Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Shabani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- Department of Anesthesiology, School of Allied Medical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Mehdi Rezaee
- Department of Anesthesiology, School of Medicine, Shahid Madani Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Zarei
- Tehran Heart Center, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
40
|
Mathebela P, Damane BP, Mulaudzi TV, Mkhize-Khwitshana ZL, Gaudji GR, Dlamini Z. Influence of the Microbiome Metagenomics and Epigenomics on Gastric Cancer. Int J Mol Sci 2022; 23:13750. [PMID: 36430229 PMCID: PMC9693604 DOI: 10.3390/ijms232213750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Gastric cancer (GC) is one of the major causes of cancer deaths worldwide. The disease is seldomly detected early and this limits treatment options. Because of its heterogeneous and complex nature, the disease remains poorly understood. The literature supports the contribution of the gut microbiome in the carcinogenesis and chemoresistance of GC. Drug resistance is the major challenge in GC therapy, occurring as a result of rewired metabolism. Metabolic rewiring stems from recurring genetic and epigenetic factors affecting cell development. The gut microbiome consists of pathogens such as H. pylori, which can foster both epigenetic alterations and mutagenesis on the host genome. Most of the bacteria implicated in GC development are Gram-negative, which makes it challenging to eradicate the disease. Gram-negative bacterium co-infections with viruses such as EBV are known as risk factors for GC. In this review, we discuss the role of microbiome-induced GC carcinogenesis. The disease risk factors associated with the presence of microorganisms and microbial dysbiosis are also discussed. In doing so, we aim to emphasize the critical role of the microbiome on cancer pathological phenotypes, and how microbiomics could serve as a potential breakthrough in determining effective GC therapeutic targets. Additionally, consideration of microbial dysbiosis in the GC classification system might aid in diagnosis and treatment decision-making, taking the specific pathogen/s involved into account.
Collapse
Affiliation(s)
- Precious Mathebela
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Hatfield 0028, South Africa
| | - Botle Precious Damane
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Hatfield 0028, South Africa
| | - Thanyani Victor Mulaudzi
- Department of Surgery, Steve Biko Academic Hospital, University of Pretoria, Hatfield 0028, South Africa
| | - Zilungile Lynette Mkhize-Khwitshana
- School of Medicine, University of Kwa-Zulu Natal, Durban, KwaZulu-Natal 4013, South Africa
- SAMRC Research Capacity Development Division, South African Medical Research Council, Tygerberg, Cape Town 7501, South Africa
| | - Guy Roger Gaudji
- Department of Urology, Level 7, Bridge C, Steve Biko Academic Hospital, Faculty of Health Sciences, University of Pretoria, Private Bag X323, Arcadia 0007, South Africa
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa
| |
Collapse
|
41
|
Davoodvandi A, Nikfar B, Reiter RJ, Asemi Z. Melatonin and cancer suppression: insights into its effects on DNA methylation. Cell Mol Biol Lett 2022; 27:73. [PMID: 36064311 PMCID: PMC9446540 DOI: 10.1186/s11658-022-00375-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022] Open
Abstract
Melatonin is an important naturally occurring hormone in mammals. Melatonin-mediated biological effects include the regulation of circadian rhythms, which is important for optimal human health. Also, melatonin has a broad range of immunoenhancing actions. Moreover, its oncostatic properties, especially regarding breast cancer, involve a variety cancer-inhibitory processes and are well documented. Due to their promising effects on the prognosis of cancer patients, anti-cancer drugs with epigenetic actions have attracted a significant amount of attention in recent years. Epigenetic modifications of cancers are categorized into three major processes including non-coding RNAs, histone modification, and DNA methylation. Hence, the modification of the latter epigenetic event is currently considered an effective strategy for treatment of cancer patients. Thereby, this report summarizes the available evidence that investigated melatonin-induced effects in altering the status of DNA methylation in different cancer cells and models, e.g., malignant glioma and breast carcinoma. Also, we discuss the role of artificial light at night (ALAN)-mediated inhibitory effects on melatonin secretion and subsequent impact on global DNA methylation of cancer cells.
Collapse
Affiliation(s)
- Amirhossein Davoodvandi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Banafsheh Nikfar
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health. Long School of Medicine, San Antonio, TX, USA
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
42
|
Novel Therapies for the Treatment of Cardiac Fibrosis Following Myocardial Infarction. Biomedicines 2022; 10:biomedicines10092178. [PMID: 36140279 PMCID: PMC9496565 DOI: 10.3390/biomedicines10092178] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 12/03/2022] Open
Abstract
Cardiac fibrosis is a common pathological consequence of most myocardial diseases. It is associated with the excessive accumulation of extracellular matrix proteins as well as fibroblast differentiation into myofibroblasts in the cardiac interstitium. This structural remodeling often results in myocardial dysfunctions such as arrhythmias and impaired systolic function in patients with heart conditions, ultimately leading to heart failure and death. An understanding of the precise mechanisms of cardiac fibrosis is still limited due to the numerous signaling pathways, cells, and mediators involved in the process. This review article will focus on the pathophysiological processes associated with the development of cardiac fibrosis. In addition, it will summarize the novel strategies for anti-fibrotic therapies such as epigenetic modifications, miRNAs, and CRISPR technologies as well as various medications in cellular and animal models.
Collapse
|
43
|
Nazeer NU, Bhat MA, Rah B, Bhat GR, Wani SI, Yousuf A, Dar AM, Afroze D. Mutational Assessment in NKX2-5 and ACTC1 Genes in Patients with Congenital Cardiac Septal Defect (CCSD) from Ethnic Kashmiri Population. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19169884. [PMID: 36011517 PMCID: PMC9408331 DOI: 10.3390/ijerph19169884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 05/10/2023]
Abstract
(1) Background globe. The etiology of CHDs is complex and involves both genetic and non-genetic factors. Although, significant progress has been made in deciphering the genetic components involved in CHDs, recent reports have revealed that mutations in Nk2 homeobox5 (NKX2-5) and actin alpha cardiac muscle1 (ACTC1) genes play a key role in CHDs such as atrial and ventricular septum defects. Therefore, the present study evaluates the role of key hotspot mutations in NKX2-5 and ACTC1 genes of congenital cardiac septal defect (CCSD) in ethnic Kashmiri population. (2) Methods: A total of 112 confirmed CHD patients were included in the current study, of which 30 patients were evaluated for mutational analysis for hotspot mutations of NKX2-5 and ACTC1 genes. The total genomic DNA was extracted from the samples (cardiac tissue/blood) and were subjected to amplification for NKX2-5 (exon 1 and 2), and ACTC1 (exon 2) genes by using PCR specific primers to analyze the hotspot mutations in respective exons. The amplified products obtained were sent to Macrogen Korea for sequencing by Sanger’s method. (3) Results: Our results confirmed that not a single mutation was found in either hotspot exon 1 and 2 of NKX2-5 and exon 2 of ACTC1 in the patients included in the current study. Interestingly, a novel synonymous nucleotide variation leading to G > C transversion (GCG > GCC) was found in exon 2 of NKX2-5 gene of CCSD patient. (4) Conclusions: The current findings demonstrated the role of NKX2-5 and ACTC1 in cardiac development. The study will provide an insight in understanding the genetic etiology and highlights the role of newly identified mutations in patients with CDS’s in ethnic Kashmiri population. In silico findings revealed amino acid changes, splice site variation and the creation of new site. Furthermore, the study warrants complete screening of genes involved in CCSDs.
Collapse
Affiliation(s)
- Nadeem Ul Nazeer
- Department of CVTS, Sheri-Kashmir Institute of Medical Sciences, Srinagar 190011, India
| | - Mohammad Akbar Bhat
- Department of CVTS, Sheri-Kashmir Institute of Medical Sciences, Srinagar 190011, India
- Correspondence: (M.A.B.); (D.A.)
| | - Bilal Rah
- Advanced Centre for Human Genetics, Sheri-Kashmir Institute of Medical Sciences, Srinagar 190011, India
| | - Gh Rasool Bhat
- Advanced Centre for Human Genetics, Sheri-Kashmir Institute of Medical Sciences, Srinagar 190011, India
| | - Shadil Ibrahim Wani
- Advanced Centre for Human Genetics, Sheri-Kashmir Institute of Medical Sciences, Srinagar 190011, India
| | - Adfar Yousuf
- Advanced Centre for Human Genetics, Sheri-Kashmir Institute of Medical Sciences, Srinagar 190011, India
| | - Abdul Majeed Dar
- Department of CVTS, Sheri-Kashmir Institute of Medical Sciences, Srinagar 190011, India
| | - Dil Afroze
- Advanced Centre for Human Genetics, Sheri-Kashmir Institute of Medical Sciences, Srinagar 190011, India
- Correspondence: (M.A.B.); (D.A.)
| |
Collapse
|
44
|
Xue H, Chen X, Yu C, Deng Y, Zhang Y, Chen S, Chen X, Chen K, Yang Y, Ling W. Gut Microbially Produced Indole-3-Propionic Acid Inhibits Atherosclerosis by Promoting Reverse Cholesterol Transport and Its Deficiency Is Causally Related to Atherosclerotic Cardiovascular Disease. Circ Res 2022; 131:404-420. [PMID: 35893593 DOI: 10.1161/circresaha.122.321253] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Accumulating evidence has shown that disorders in the gut microbiota and derived metabolites affect the development of atherosclerotic cardiovascular disease (ASCVD). However, which and how specific gut microbial metabolites contribute to the progression of atherosclerosis and the clinical relevance of their alterations remain unclear. METHODS We performed integrated microbiome-metabolome analysis of 30 patients with coronary artery disease (CAD) and 30 age- and sex-matched healthy controls to identify CAD-associated microbial metabolites, which were then assessed in an independent population of patients with ASCVD and controls (n=256). We further investigate the effect of CAD-associated microbial metabolites on atherosclerosis and the mechanisms of the action. RESULTS Indole-3-propionic acid (IPA), a solely microbially derived tryptophan metabolite, was the most downregulated metabolite in patients with CAD. Circulating IPA was then shown in an independent population to be associated with risk of prevalent ASCVD and correlated with the ASCVD severity. Dietary IPA supplementation alleviates atherosclerotic plaque development in ApoE-/- mice. In murine- and human-derived macrophages, administration of IPA promoted cholesterol efflux from macrophages to ApoA-I through an undescribed miR-142-5p/ABCA1 (ATP-binding cassette transporter A1) signaling pathway. Further in vivo studies demonstrated that IPA facilitates macrophage reverse cholesterol transport, correlating with the regulation of miR-142-5p/ABCA1 pathway, whereas reduced IPA production contributed to the aberrant overexpression of miR-142-5p in macrophages and accelerated the progression of atherosclerosis. Moreover, the miR-142-5p/ABCA1/reverse cholesterol transport axis in macrophages were dysregulated in patients with CAD, and correlated with the changes in circulating IPA levels. CONCLUSIONS Our study identify a previously unknown link between specific gut microbiota-derived tryptophan metabolite and ASCVD. The microbial metabolite IPA/miR-142-5p/ABCA1 pathway may represent a promising therapeutic target for ASCVD.
Collapse
Affiliation(s)
- Hongliang Xue
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, China (H.X., Y.Y., W.L.).,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China (H.X., X.C., S.C., Y.Y., W.L.)
| | - Xu Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China (H.X., X.C., S.C., Y.Y., W.L.).,Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder (Xu Chen)
| | - Chao Yu
- Center for Health Examination, the 3 Affiliated Hospital, Sun Yat-sen University, Guangzhou, China (C.Y.)
| | - Yuqing Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Centre, Sun Yat-sen University, Guangzhou, China (Y.D.)
| | - Yuan Zhang
- Department of Geriatrics, The Third Affiliated Hospital of Guangzhou Medical University, China (Y.Z.).,Department of Cardiology, General Hospital of Guangzhou Military Command of People's Liberation Army, China (Y.Z.)
| | - Shen Chen
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China (H.X., X.C., S.C., Y.Y., W.L.)
| | - Xuechen Chen
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany (Xuechen Chen)
| | - Ke Chen
- Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China (K.C.)
| | - Yan Yang
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, China (H.X., Y.Y., W.L.).,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China (H.X., X.C., S.C., Y.Y., W.L.).,Department of Nutrition, School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, China (Y.Y.)
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, China (H.X., Y.Y., W.L.).,Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, China (H.X., X.C., S.C., Y.Y., W.L.)
| |
Collapse
|
45
|
Kaul U, Scher C, Henderson CR, Kim P, Dyhrberg M, Rudin V, Lytle M, Bundy N, Reid MC. A mobile health + health coaching application for the management of chronic non-cancer pain in older adults: Results from a pilot randomized controlled study. FRONTIERS IN PAIN RESEARCH 2022; 3:921428. [PMID: 35959237 PMCID: PMC9362151 DOI: 10.3389/fpain.2022.921428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction The rapid growth of mobile health (mHealth) devices holds substantial potential for improving care and care outcomes in aging adults with chronic non-cancer pain (CNCP), however, research evaluating these devices in older adults remains limited. Objective To ascertain the feasibility and preliminary efficacy of an mHealth intervention (Mymee) that combines symptom, diet, and behavior tracking via a smartphone application with data analytics to detect associations between symptoms and lifestyle factors along with weekly health coaching sessions to mitigate CNCP in adults 55 years of age and older. Methods Participants (N = 31) in this pilot study were recruited from one primary care practice in New York City and randomized to an intervention [app + up to 12 health coaching sessions (scheduled approximately once weekly) + usual care] or a control (app + usual care) arm. Feasibility measures included recruitment (proportion of eligible persons who enrolled) and retention rates (proportion of subjects completing a follow-up assessment) as well as adherence with the weekly coaching sessions and logging daily data on the app. Efficacy outcomes (e.g., pain intensity, self-efficacy, disability, anxiety) were assessed at baseline and follow-up (~16 weeks after baseline). Descriptive statistics were obtained and general linear mixed models used for primary analyses. Results Participants had a mean (standard deviation) age of 67.32 (9.17) and were mostly female (61%). Feasibility outcomes were mixed as evidenced by recruitment and retention rates of 74% and 65%, respectively. The mean number of weekly coaching sessions attended by intervention participants was 6.05 (SD = 5.35), while the average number of days logging data on the app was 44.82 (34.02). We found a consistent trend in favor of the intervention, where pain intensity, affect, and quality of life measures improved considerably more among intervention (vs. control) participants. Finally, the proportion of participants with GAD-7 scores at follow up decreased by 0.35 to 0, whereas controls did not change, a significant effect in favor of the intervention (p = 0.02). Conclusions This study supports the need for future research that seeks to enhance feasibility outcomes and confirm the efficacy of the Mymee intervention among aging adults with CNCP.
Collapse
Affiliation(s)
- Usha Kaul
- Division of Geriatrics and Palliative Medicine, Weill Cornell Medical Center, New York, NY, United States
| | - Clara Scher
- Rutgers School of Social Work, New Brunswick, NJ, United States
| | | | - Patricia Kim
- Division of Geriatrics and Palliative Medicine, Weill Cornell Medical Center, New York, NY, United States
| | | | | | | | | | - M. Carrington Reid
- Division of Geriatrics and Palliative Medicine, Weill Cornell Medical Center, New York, NY, United States
| |
Collapse
|
46
|
Li L, Xu N, Liu J, Chen Z, Liu X, Wang J. m6A Methylation in Cardiovascular Diseases: From Mechanisms to Therapeutic Potential. Front Genet 2022; 13:908976. [PMID: 35836571 PMCID: PMC9274458 DOI: 10.3389/fgene.2022.908976] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 06/07/2022] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide. Recent studies have shown that n6-methyladenosine (m6A) plays a major role in cardiovascular homeostasis and pathophysiology. These studies have confirmed that m6A methylation affects the pathophysiology of cardiovascular diseases by regulating cellular processes such as differentiation, proliferation, inflammation, autophagy, and apoptosis. Moreover, plenty of research has confirmed that m6A modification can delay the progression of CVD via the post-transcriptional regulation of RNA. However, there are few available summaries of m6A modification regarding CVD. In this review, we highlight advances in CVD-specific research concerning m6A modification, summarize the mechanisms underlying the involvement of m6A modification during the development of CVD, and discuss the potential of m6A modification as a therapeutic target of CVD.
Collapse
Affiliation(s)
| | | | | | | | | | - Junnan Wang
- Department of Cardiology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
47
|
The Role of Epigenetic Modifications in Late Complications in Type 1 Diabetes. Genes (Basel) 2022; 13:genes13040705. [PMID: 35456511 PMCID: PMC9029845 DOI: 10.3390/genes13040705] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 11/29/2022] Open
Abstract
Type 1 diabetes is a chronic autoimmune disease in which the destruction of pancreatic β cells leads to hyperglycemia. The prevention of hyperglycemia is very important to avoid or at least postpone the development of micro- and macrovascular complications, also known as late complications. These include diabetic retinopathy, chronic renal failure, diabetic neuropathy, and cardiovascular diseases. The impact of long-term hyperglycemia has been shown to persist long after the normalization of blood glucose levels, a phenomenon known as metabolic memory. It is believed that epigenetic mechanisms such as DNA methylation, histone modifications, and microRNAs, play an important role in metabolic memory. The aim of this review is to address the impact of long-term hyperglycemia on epigenetic marks in late complications of type 1 diabetes.
Collapse
|
48
|
Pei S, Zhao H, Chen L, He X, Hua Q, Meng X, Shi R, Zhang J, Zhang H, Liu R, Li D. Preventive Effect of Ellagic Acid on Cardiac Dysfunction in Diabetic Mice through Regulating DNA Hydroxymethylation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:1902-1910. [PMID: 35129965 DOI: 10.1021/acs.jafc.1c07574] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Ellagic acid (EA) improves mitochondrial dysfunction and protects diabetic hearts. The mitochondrial tricarboxylic acid (TCA) cycle regulates DNA 5-hydroxymethylcytosine (5hmC) levels by affecting activity of 10-11 translocation enzymes (TETs). Therefore, we hypothesized that EA prevents diabetic cardiac dysfunction by modulating DNA 5hmC levels. C57BL/6J mice were fed a high-fat diet to induce diabetes and treated with EA (100 mg kg-1 day-1) for 8 weeks. Serum concentrations of glucose, insulin, and triglyceride and aspartate transaminase and creatine kinase activities were significantly lower in the EA group than the diabetes mellitus (DM) group. DNA 5hmC levels of mice hearts were significantly higher in the EA group than the DM group. The protein levels of TET, complexes I/III/V were significantly higher in the EA group than the DM group. The results shows that EA has a preventive effect on diabetic cardiac dysfunction, which may be achieved by upregulating TET activity through improving the TCA cycle, to reshape DNA 5hmC levels of mice hearts.
Collapse
Affiliation(s)
- Shengjie Pei
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
- School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Haifeng Zhao
- Qingdao Institute for Food and Drug Control, Qingdao, Shandong 266071, People's Republic of China
| | - Lei Chen
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
- School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Xin He
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
- School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Qinglian Hua
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
- School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Xiangyuan Meng
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
- School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Ruiqing Shi
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
- School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Jingyuan Zhang
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
- School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Hong Zhang
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Run Liu
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
- School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| | - Duo Li
- Institute of Nutrition & Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
- School of Public Health, Qingdao University, Qingdao, Shandong 266071, People's Republic of China
| |
Collapse
|
49
|
Jung M, Ahn YS, Chang SJ, Kim CB, Jeong KS, Koh SB, Gim JA. Variation in Genotype and DNA Methylation Patterns Based on Alcohol Use and CVD in the Korean Genome and Epidemiology Study (KoGES). Genes (Basel) 2022; 13:genes13020172. [PMID: 35205218 PMCID: PMC8871634 DOI: 10.3390/genes13020172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/09/2022] [Accepted: 01/14/2022] [Indexed: 12/14/2022] Open
Abstract
Alcohol consumption can increase the risk of chronic diseases, such as myocardial infarction, coronary artery disease, hyperlipidemia, and hypertension. We aimed to assess the association between genotype, DNA methylation patterns, alcohol consumption, and chronic diseases in Korean population. We analyzed 8840 subjects for genotypes and 446 for DNA methylation among the 9351 subjects from the Korean Genome and Epidemiology Study (KoGES). We further divided both groups into two sub-groups according to the presence/absence of chronic diseases. We selected genes whose methylation varied significantly with alcohol consumption, and visualized genotype and DNA methylation patterns specific to each group. Genome-wide association study (GWAS) revealed single nucleotide polymorphisms (SNPs) rs2074356 and rs11066280 in HECT domain E3 ubiquitin protein ligase 4 (HECTD4) to be significantly associated with alcohol consumption in both the presence. The rs12229654 genotype also displayed significantly different patterns with alcohol consumption. Furthermore, we retrieved differentially methylated regions (DMRs) from four groups based on sex and chronic diseases and compared them by drinking status. In genotype analysis, cardiovascular diseases (CVDs) showed a higher proportion in drinker than in non-drinker, but not in DMR analysis. Additionally, we analyzed the enriched Gene Ontology terms and Kyoto Gene and Genome Encyclopedia (KEGG) pathways and visualized the network, heatmap, and upset plot. We show that the pattern of DNA methylation associated with CVD is strongly influenced by alcoholism. Overall, this study identified genetic and epigenetic variants influenced by alcohol consumption and chronic diseases.
Collapse
Affiliation(s)
- Myoungjee Jung
- Department of Preventive Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Korea; (M.J.); (S.-J.C.); (C.-B.K.)
| | - Yeon-Soon Ahn
- Department of Preventive Medicine and Genomic Cohort Institute, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Korea;
| | - Sei-Jin Chang
- Department of Preventive Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Korea; (M.J.); (S.-J.C.); (C.-B.K.)
| | - Chun-Bae Kim
- Department of Preventive Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Korea; (M.J.); (S.-J.C.); (C.-B.K.)
| | - Kyoung Sook Jeong
- Department of Occupational and Environmental Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Korea;
| | - Sang-Baek Koh
- Department of Preventive Medicine, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Korea; (M.J.); (S.-J.C.); (C.-B.K.)
- Institute of Genomic Cohort, Yonsei University Wonju College of Medicine, 20 Ilsan-ro, Wonju 26426, Korea
- Correspondence: (S.-B.K.); (J.-A.G.); Tel.: +82-33-741-0345 (S.-B.K.); +82-2-2626-2362 (J.-A.G.)
| | - Jeong-An Gim
- Medical Science Research Center, College of Medicine, Korea University, 8 Gamasan-ro 20-gil, Guro-gu, Seoul 08308, Korea
- Correspondence: (S.-B.K.); (J.-A.G.); Tel.: +82-33-741-0345 (S.-B.K.); +82-2-2626-2362 (J.-A.G.)
| |
Collapse
|
50
|
Chen J, Liu Z, Ma L, Gao S, Fu H, Wang C, Lu A, Wang B, Gu X. Targeting Epigenetics and Non-coding RNAs in Myocardial Infarction: From Mechanisms to Therapeutics. Front Genet 2022; 12:780649. [PMID: 34987550 PMCID: PMC8721121 DOI: 10.3389/fgene.2021.780649] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Myocardial infarction (MI) is a complicated pathology triggered by numerous environmental and genetic factors. Understanding the effect of epigenetic regulation mechanisms on the cardiovascular disease would advance the field and promote prophylactic methods targeting epigenetic mechanisms. Genetic screening guides individualised MI therapies and surveillance. The present review reported the latest development on the epigenetic regulation of MI in terms of DNA methylation, histone modifications, and microRNA-dependent MI mechanisms and the novel therapies based on epigenetics.
Collapse
Affiliation(s)
- Jinhong Chen
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Zhichao Liu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Li Ma
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Shengwei Gao
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Huanjie Fu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Can Wang
- Acupuncture Department, The First Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Anmin Lu
- Department of TCM, Tianjin University of TCM, Tianjin, China
| | - Baohe Wang
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| | - Xufang Gu
- Department of Cardiology, The Second Affiliated Hospital of Tianjin University of TCM, Tianjin, China
| |
Collapse
|