1
|
da Silva Rodrigues Marçal E, Borges JB, Bastos GM, Crespo Hirata TD, de Oliveira VF, Gonçalves RM, Faludi AA, Dias França JI, de Oliveira Silva DV, Malaquias VB, Luchessi AD, Silbiger VN, Nakazone MA, Carmo TS, Silva Souza DR, Sampaio MF, Crespo Hirata RD, Hirata MH. Methylation status of LDLR, PCSK9 and LDLRAP1 is associated with cardiovascular events in familial hypercholesterolemia. Epigenomics 2024; 16:809-820. [PMID: 38884343 PMCID: PMC11370914 DOI: 10.1080/17501911.2024.2351792] [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: 02/26/2024] [Accepted: 04/27/2024] [Indexed: 06/18/2024] Open
Abstract
Aim: Methylation of LDLR, PCSK9 and LDLRAP1 CpG sites was assessed in patients with familial hypercholesterolemia (FH). Methods: DNA methylation of was analyzed by pyrosequencing in 131 FH patients and 23 normolipidemic (NL) subjects.Results: LDLR, PCSK9 and LDLRP1 methylation was similar between FH patients positive (MD) and negative (non-MD) for pathogenic variants in FH-related genes. LDLR and PCSK9 methylation was higher in MD and non-MD groups than NL subjects (p < 0.05). LDLR, PCSK9 and LDLRAP1 methylation profiles were associated with clinical manifestations and cardiovascular events in FH patients (p < 0.05).Conclusion: Differential methylation of LDLR, PCSK9 and LDLRAP1 is associated with hypercholesterolemia and cardiovascular events. This methylation profile maybe useful as a biomarker and contribute to the management of FH.
Collapse
Affiliation(s)
- Elisangela da Silva Rodrigues Marçal
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
- Laboratory of Molecular Research in Cardiology, Institute of Cardiology Dante Pazzanese, Sao Paulo, 04012-909, Brazil
| | - Jéssica Bassani Borges
- Department of Research, Hospital Beneficiencia Portuguesa de Sao Paulo, Sao Paulo, 01323-001, Brazil
| | - Gisele Medeiros Bastos
- Department of Research, Hospital Beneficiencia Portuguesa de Sao Paulo, Sao Paulo, 01323-001, Brazil
| | - Thiago Dominguez Crespo Hirata
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Victor Fernandes de Oliveira
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | | | - Andre Arpad Faludi
- Medical Clinic Division, Institute of Cardiology Dante Pazzanese, Sao Paulo, 04012-909, Brazil
| | - João Italo Dias França
- Center for Clinical Trials & Pharmacovigilance, Butantan Institute, Sao Paulo, 05585-000, Brazil
| | - Daiana Vitor de Oliveira Silva
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Vanessa Barbosa Malaquias
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Andre Ducati Luchessi
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Natal, 59012-570, Brazil
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Natal, 59012-570, Brazil
| | - Vivian Nogueira Silbiger
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Natal, 59012-570, Brazil
- Graduate Program in Pharmaceutical Sciences, Federal University of Rio Grande do Norte, Natal, 59012-570, Brazil
| | - Marcelo Arruda Nakazone
- Department of Cardiology & Cardiovascular Surgery, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto, 15090-000, Brazil
| | - Tayanne Silva Carmo
- Department of Biochemistry & Molecular Biology, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto, 15090-000, Brazil
| | - Dorotéia Rossi Silva Souza
- Department of Biochemistry & Molecular Biology, Sao Jose do Rio Preto Medical School, Sao Jose do Rio Preto, 15090-000, Brazil
| | - Marcelo Ferraz Sampaio
- Department of Cardiology, Hospital Beneficencia Portuguesa de Sao Paulo, Sao Paulo, 01323-001, Brazil
| | - Rosario Dominguez Crespo Hirata
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| | - Mario Hiroyuki Hirata
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, 05508-000, Brazil
| |
Collapse
|
2
|
Infante T, Pepin ME, Ruocco A, Trama U, Mauro C, Napoli C. CDK5R1, GSE1, HSPG2 and WDFY3 as indirect epigenetic-sensitive genes in atrial fibrillation. Eur J Clin Invest 2024; 54:e14135. [PMID: 37991085 DOI: 10.1111/eci.14135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND Although mounting evidence supports that aberrant DNA methylation occurs in the hearts of patients with atrial fibrillation (AF), noninvasive epigenetic characterization of AF has not yet been defined. METHODS We investigated DNA methylome changes in peripheral blood CD4+ T cells isolated from 10 patients with AF relative to 11 healthy subjects (HS) who were enrolled in the DIANA clinical trial (NCT04371809) via reduced-representation bisulfite sequencing (RRBS). RESULTS An atrial-specific PPI network revealed 18 hub differentially methylated genes (DMGs), wherein ROC curve analysis revealed reasonable diagnostic performance of DNA methylation levels found within CDK5R1 (AUC = 0.76; p = 0.049), HSPG2 (AUC = 0.77; p = 0.038), WDFY3 (AUC = 0.78; p = 0.029), USP49 (AUC = 0.76; p = 0.049), GSE1 (AUC = 0.76; p = 0.049), AIFM1 (AUC = 0.76; p = 0.041), CDK5RAP2 (AUC = 0.81; p = 0.017), COL4A1 (AUC = 0.86; p < 0.001), SEPT8 (AUC = 0.90; p < 0.001), PFDN1 (AUC = 0.90; p < 0.01) and ACOT7 (AUC = 0.78; p = 0.032). Transcriptional profiling of the hub DMGs provided a significant overexpression of PSDM6 (p = 0.004), TFRC (p = 0.01), CDK5R1 (p < 0.001), HSPG2 (p = 0.01), WDFY3 (p < 0.001), USP49 (p = 0.004) and GSE1 (p = 0.021) in AF patients vs HS. CONCLUSIONS CDK5R1, GSE1, HSPG2 and WDFY3 resulted the best discriminatory genes both at methylation and gene expression level. Our results provide several candidate diagnostic biomarkers with the potential to advance precision medicine in AF.
Collapse
Affiliation(s)
- Teresa Infante
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mark E Pepin
- Division of Internal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- The Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
| | - Antonio Ruocco
- Cardiology Division, "A. Cardarelli" Hospital, Naples, Italy
| | - Ugo Trama
- General Direction of Health Care & Regional Health System Coordination, Drug & Device Politics, Campania Region, Naples, Italy
| | - Ciro Mauro
- Cardiology Division, "A. Cardarelli" Hospital, Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
3
|
Patil NS, Larocque N, van der Pol CB, Torres C, Raptis DA, Patlas MN. Chemotherapy-Induced Toxicities: An Imaging Primer. Can Assoc Radiol J 2023; 74:432-445. [PMID: 35968850 DOI: 10.1177/08465371221120263] [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] [Indexed: 01/03/2023] Open
Abstract
The Coronavirus Disease of 2019 (COVID-19) pandemic has caused significant delays in the delivery of cancer treatments in Canada. As cancer treatment and imaging volumes return to normal, radiologists will encounter more cases of chemotherapy-induced toxicities. These toxicities have varied appearances on imaging, and can affect multiple organ systems. The purpose of this review is to offer a unified resource for general radiologists regarding the imaging appearances of chemotherapy-induced toxicities.
Collapse
Affiliation(s)
- Nikhil S Patil
- Michael DeGroote School of Medicine, 62703McMaster University, Hamilton, ON, Canada
| | - Natasha Larocque
- Department of Radiology, 3710McMaster University, Hamilton, ON, Canada
| | - Christian B van der Pol
- Department of Diagnostic Imaging, Juravinski Hospital and Cancer Centre, Hamilton Health Sciences, Canada
| | - Carlos Torres
- Department of Radiology, Radiation Oncology and Medical Physics, 6363University of Ottawa, Ottawa, ON, Canada
| | - Demetrios A Raptis
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA
| | - Michael N Patlas
- Department of Radiology, 3710McMaster University, Hamilton, ON, Canada
| |
Collapse
|
4
|
Schiano C, Balbi C, de Nigris F, Napoli C. Basic Pathogenic Mechanisms and Epigenetic Players Promoted by Extracellular Vesicles in Vascular Damage. Int J Mol Sci 2023; 24:ijms24087509. [PMID: 37108672 PMCID: PMC10138986 DOI: 10.3390/ijms24087509] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/14/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
Both progression from the early pathogenic events to clinically manifest cardiovascular diseases (CVD) and cancer impact the integrity of the vascular system. Pathological vascular modifications are affected by interplay between endothelial cells and their microenvironment. Soluble factors, extracellular matrix molecules and extracellular vesicles (EVs) are emerging determinants of this network that trigger specific signals in target cells. EVs have gained attention as package of molecules with epigenetic reversible activity causing functional vascular changes, but their mechanisms are not well understood. Valuable insights have been provided by recent clinical studies, including the investigation of EVs as potential biomarkers of these diseases. In this paper, we review the role and the mechanism of exosomal epigenetic molecules during the vascular remodeling in coronary heart disease as well as in cancer-associated neoangiogenesis.
Collapse
Affiliation(s)
- Concetta Schiano
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Institute, 6807 Taverne-Torricella, Switzerland
| | - Carolina Balbi
- Laboratory of Cellular and Molecular Cardiology, Cardiocentro Ticino Institute, 6807 Taverne-Torricella, Switzerland
| | - Filomena de Nigris
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania Luigi Vanvitelli, 80138 Naples, Italy
- Clinical Department of Internal Medicine and Specialistic Units, Division of Clinical Immunology and Immunohematology, Transfusion Medicine and Transplant Immunology (SIMT), Azienda Universitaria Policlinico (AOU), 80138 Naples, Italy
| |
Collapse
|
5
|
Schiano C, Balbi C, Burrello J, Ruocco A, Infante T, Fiorito C, Panella S, Barile L, Mauro C, Vassalli G, Napoli C. De novo DNA methylation induced by circulating extracellular vesicles from acute coronary syndrome patients. Atherosclerosis 2022; 354:41-52. [PMID: 35830762 DOI: 10.1016/j.atherosclerosis.2022.06.1026] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/25/2022] [Accepted: 06/29/2022] [Indexed: 11/02/2022]
Abstract
BACKGROUND AND AIMS DNA methylation is associated with gene silencing, but its clinical role in cardiovascular diseases (CVDs) remains to be elucidated. We hypothesized that extracellular vesicles (EVs) may carry epigenetic changes, showing themselves as a potentially valuable non-invasive diagnostic liquid biopsy. We isolated and characterized circulating EVs of acute coronary syndrome (ACS) patients and assessed their role on DNA methylation in epigenetic modifications. METHODS EVs were recovered from plasma of 19 ACS patients and 50 healthy subjects (HS). Flow cytometry, qRT-PCR, and Western blot (WB) were performed to evaluate both intra-vesicular and intra-cellular signals. ShinyGO, PANTHER, and STRING tools were used to perform GO and PPI network analyses. RESULTS ACS-derived EVs showed increased levels of DNA methyltransferases (DNMTs) (p<0.001) and Ten-eleven translocation (TET) genes reduction. Specifically, de novo methylation transcripts, as DNMT3A and DNMT3B, were significantly increased in plasma ACS-EVs. DNA methylation analysis on PBMCs from healthy donors treated with HS- and ACS-derived EVs showed an important role of DNMTs carried by EVs. PPI network analysis evidenced that ACS-EVs induced changes in PBMC methylome. In the most enriched subnetwork, the hub gene SRC was connected to NOTCH1, FOXO3, CDC42, IKBKG, RXRA, DGKG, BAIAP2 genes that were showed to have many molecular effects on various cell types into onset of several CVDs. Modulation in gene expression after ACS-EVs treatment was confirmed for SRC, NOTCH1, FOXO3, RXRA, DGKG and BAIAP2 (p<0.05). CONCLUSIONS Our data showed an important role for ACS-derived EVs in gene expression modulation through de novo DNA methylation signals, and modulating signalling pathways in target cells.
Collapse
Affiliation(s)
- Concetta Schiano
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania Luigi Vanvitelli, Naples, Italy; Cellular and Molecular Cardiology lab Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland; Laboratories for Translation Research, EOC, Bellinzona, Switzerland.
| | - Carolina Balbi
- Cellular and Molecular Cardiology lab Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland; Laboratories for Translation Research, EOC, Bellinzona, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| | - Jacopo Burrello
- Laboratories for Translation Research, EOC, Bellinzona, Switzerland; Cardiovascular Theranostics, Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland
| | - Antonio Ruocco
- Unit of Cardiovascular Diseases and Arrhythmias, Antonio Cardarelli Hospital, Naples, Italy
| | - Teresa Infante
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Stefano Panella
- Laboratories for Translation Research, EOC, Bellinzona, Switzerland; Cardiovascular Theranostics, Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland
| | - Lucio Barile
- Laboratories for Translation Research, EOC, Bellinzona, Switzerland; Cardiovascular Theranostics, Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland
| | - Ciro Mauro
- Unit of Cardiovascular Diseases and Arrhythmias, Antonio Cardarelli Hospital, Naples, Italy
| | - Giuseppe Vassalli
- Cellular and Molecular Cardiology lab Istituto Cardiocentro Ticino-EOC, Lugano, Switzerland; Laboratories for Translation Research, EOC, Bellinzona, Switzerland; Center for Molecular Cardiology, Zurich, Switzerland
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania Luigi Vanvitelli, Naples, Italy; Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology (SIMT), Regional Reference Laboratory of Transplant Immunology (LIT), Azienda Universitaria Policlinico (AOU), Naples, Italy
| |
Collapse
|
6
|
Galow AM, Peleg S. How to Slow down the Ticking Clock: Age-Associated Epigenetic Alterations and Related Interventions to Extend Life Span. Cells 2022; 11:468. [PMID: 35159278 PMCID: PMC8915189 DOI: 10.3390/cells11030468] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 01/26/2022] [Indexed: 02/04/2023] Open
Abstract
Epigenetic alterations pose one major hallmark of organismal aging. Here, we provide an overview on recent findings describing the epigenetic changes that arise during aging and in related maladies such as neurodegeneration and cancer. Specifically, we focus on alterations of histone modifications and DNA methylation and illustrate the link with metabolic pathways. Age-related epigenetic, transcriptional and metabolic deregulations are highly interconnected, which renders dissociating cause and effect complicated. However, growing amounts of evidence support the notion that aging is not only accompanied by epigenetic alterations, but also at least in part induced by those. DNA methylation clocks emerged as a tool to objectively determine biological aging and turned out as a valuable source in search of factors positively and negatively impacting human life span. Moreover, specific epigenetic signatures can be used as biomarkers for age-associated disorders or even as targets for therapeutic approaches, as will be covered in this review. Finally, we summarize recent potential intervention strategies that target epigenetic mechanisms to extend healthy life span and provide an outlook on future developments in the field of longevity research.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute for Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Shahaf Peleg
- Research Group Epigenetics, Metabolism and Longevity, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
- Institute of Neuroregeneration and Neurorehabilitation of Qingdao University, Qingdao 266071, China
| |
Collapse
|
7
|
Infante T, Cavaliere C, Punzo B, Grimaldi V, Salvatore M, Napoli C. Radiogenomics and Artificial Intelligence Approaches Applied to Cardiac Computed Tomography Angiography and Cardiac Magnetic Resonance for Precision Medicine in Coronary Heart Disease: A Systematic Review. Circ Cardiovasc Imaging 2021; 14:1133-1146. [PMID: 34915726 DOI: 10.1161/circimaging.121.013025] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The risk of coronary heart disease (CHD) clinical manifestations and patient management is estimated according to risk scores accounting multifactorial risk factors, thus failing to cover the individual cardiovascular risk. Technological improvements in the field of medical imaging, in particular, in cardiac computed tomography angiography and cardiac magnetic resonance protocols, laid the development of radiogenomics. Radiogenomics aims to integrate a huge number of imaging features and molecular profiles to identify optimal radiomic/biomarker signatures. In addition, supervised and unsupervised artificial intelligence algorithms have the potential to combine different layers of data (imaging parameters and features, clinical variables and biomarkers) and elaborate complex and specific CHD risk models allowing more accurate diagnosis and reliable prognosis prediction. Literature from the past 5 years was systematically collected from PubMed and Scopus databases, and 60 studies were selected. We speculated the applicability of radiogenomics and artificial intelligence through the application of machine learning algorithms to identify CHD and characterize atherosclerotic lesions and myocardial abnormalities. Radiomic features extracted by cardiac computed tomography angiography and cardiac magnetic resonance showed good diagnostic accuracy for the identification of coronary plaques and myocardium structure; on the other hand, few studies exploited radiogenomics integration, thus suggesting further research efforts in this field. Cardiac computed tomography angiography resulted the most used noninvasive imaging modality for artificial intelligence applications. Several studies provided high performance for CHD diagnosis, classification, and prognostic assessment even though several efforts are still needed to validate and standardize algorithms for CHD patient routine according to good medical practice.
Collapse
Affiliation(s)
- Teresa Infante
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy (T.I., C.N.)
| | | | - Bruna Punzo
- IRCCS SDN, Naples, Italy (C.C., B.P., V.G., M.S., C.N.)
| | | | | | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy (T.I., C.N.).,IRCCS SDN, Naples, Italy (C.C., B.P., V.G., M.S., C.N.)
| |
Collapse
|
8
|
Machine Learning and Bioinformatics Framework Integration to Potential Familial DCM-Related Markers Discovery. Genes (Basel) 2021; 12:genes12121946. [PMID: 34946895 PMCID: PMC8701745 DOI: 10.3390/genes12121946] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/11/2022] Open
Abstract
Objectives: Dilated cardiomyopathy (DCM) is characterized by a specific transcriptome. Since the DCM molecular network is largely unknown, the aim was to identify specific disease-related molecular targets combining an original machine learning (ML) approach with protein-protein interaction network. Methods: The transcriptomic profiles of human myocardial tissues were investigated integrating an original computational approach, based on the Custom Decision Tree algorithm, in a differential expression bioinformatic framework. Validation was performed by quantitative real-time PCR. Results: Our preliminary study, using samples from transplanted tissues, allowed the discovery of specific DCM-related genes, including MYH6, NPPA, MT-RNR1 and NEAT1, already known to be involved in cardiomyopathies Interestingly, a combination of these expression profiles with clinical characteristics showed a significant association between NEAT1 and left ventricular end-diastolic diameter (LVEDD) (Rho = 0.73, p = 0.05), according to severity classification (NYHA-class III). Conclusions: The use of the ML approach was useful to discover preliminary specific genes that could lead to a rapid selection of molecular targets correlated with DCM clinical parameters. For the first time, NEAT1 under-expression was significantly associated with LVEDD in the human heart.
Collapse
|
9
|
Miroshnikova VV, Panteleeva AA, Pobozheva IA, Razgildina ND, Polyakova EA, Markov AV, Belyaeva OD, Berkovich OA, Baranova EI, Nazarenko MS, Puzyrev VP, Pchelina SN. ABCA1 and ABCG1 DNA methylation in epicardial adipose tissue of patients with coronary artery disease. BMC Cardiovasc Disord 2021; 21:566. [PMID: 34837967 PMCID: PMC8627066 DOI: 10.1186/s12872-021-02379-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Recent studies have focused on the potential role of epicardial adipose tissue (EAT) in the development of coronary artery disease (CAD). ABCA1 and ABCG1 transporters regulate cell cholesterol content and reverse cholesterol transport. We aimed to determine whether DNA methylation and mRNA levels of the ABCA1 and ABCG1 genes in EAT and subcutaneous adipose tissue (SAT) were associated with CAD. METHODS Paired EAT and SAT samples were collected from 82 patients undergoing elective cardiac surgery either for coronary artery bypass grafting (CAD group, N = 66) or valve surgery (NCAD group, N = 16). ABCA1 and ABCG1 mRNA levels in EAT and SAT samples were analyzed using real time polymerase chain reaction, ABCA1 protein levels in EAT samples were assessed by western blotting. ABCA1 and ABCG1 DNA methylation analysis was performed in 24 samples from the CAD group and 9 samples from the NCAD group via pyrosequencing. RESULTS DNA methylation levels in the ABCA1 promoter and ABCG1 cg27243685 and cg06500161 CpG sites were higher in EAT samples from patients with CAD compared with NCAD (21.92% vs 10.81%, p = 0.003; 71.51% vs 68.42%, p = 0.024; 46.11% vs 37.79%, p = 0.016, respectively). In patients with CAD, ABCA1 and ABCG1 DNA methylation levels were higher in EAT than in SAT samples (p < 0.05). ABCA1 mRNA levels in EAT samples were reduced in the subgroup of patients with CAD and concomitant carotid artery disease or peripheral artery disease compared with the NCAD group (p = 0.024). ABCA1 protein levels in EAT samples tended to be lower in CAD patients than in the NCAD group (p = 0.053). DNA methylation levels at the ABCG1 cg27243685 site positively correlated with plasma triglyceride concentration (r = 0.510, p = 0.008), body mass index (r = 0.556, p = 0.013) and waist-to-hip ratio (r = 0.504, p = 0.012) in SAT samples. CONCLUSION CAD is associated with ABCA1 and ABCG1 DNA hypermethylation in EAT. CAD with concomitant carotid artery disease or peripheral artery disease is accompanied by decreased ABCA1 gene expression in EAT. DNA methylation levels at the ABCG1 cg27243685 locus in SAT are associated with hypertriglyceridemia and obesity.
Collapse
Affiliation(s)
- Valentina V Miroshnikova
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation.
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation.
| | - Alexandra A Panteleeva
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
- National Research Centre "Kurchatov Institute", Moscow, Russia
| | - Irina A Pobozheva
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
- National Research Centre "Kurchatov Institute", Moscow, Russia
| | - Natalia D Razgildina
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
| | - Ekaterina A Polyakova
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Anton V Markov
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk, Russian Federation
| | - Olga D Belyaeva
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Olga A Berkovich
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Elena I Baranova
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
| | - Maria S Nazarenko
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk, Russian Federation
| | - Valery P Puzyrev
- Laboratory of Population Genetics, Research Institute of Medical Genetics, Tomsk, Russian Federation
| | - Sofya N Pchelina
- Petersburg Nuclear Physics Institute Named By B.P. Konstantinov of National Research Center "Kurchatov Institute", Gatchina, Russian Federation
- Pavlov First Saint Petersburg State Medical University, St.-Petersburg, Russian Federation
- National Research Centre "Kurchatov Institute", Moscow, Russia
| |
Collapse
|
10
|
Montisci A, Palmieri V, Liu JE, Vietri MT, Cirri S, Donatelli F, Napoli C. Severe Cardiac Toxicity Induced by Cancer Therapies Requiring Intensive Care Unit Admission. Front Cardiovasc Med 2021; 8:713694. [PMID: 34540917 PMCID: PMC8446380 DOI: 10.3389/fcvm.2021.713694] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 07/27/2021] [Indexed: 12/28/2022] Open
Abstract
A steadying increase of cancer survivors has been observed as a consequence of more effective therapies. However, chemotherapy regimens are often associated with significant toxicity, and cardiac damage emerges as a prominent clinical issue. Many mechanisms sustain chemotherapy-induced cardiac toxicity: direct myocyte damage, arrhythmia induction, coronary vasospasm, and accelerated atherosclerosis. Anthracyclines are the most studied cardiotoxic drugs and represent a clinical model for cardiac damage induced by chemotherapy. In patients suffering from advanced heart failure (HF) because of chemotherapy-related cardiomyopathy, when refractory to optimal medical therapy, mechanical circulatory support or heart transplantation represents an effective treatment. Here, the main mechanisms of cardiac toxicity induced by cancer therapies are analyzed, with a focus on patients requiring intensive care unit (ICU) admission during the course of the disease because of acute cardiac toxicity, takotsubo syndrome, and acute-on-chronic HF in patients suffering from chemotherapy-induced cardiomyopathy. In a subset of patients, cardiac toxicity can be acute and life-threatening, leading to overt cardiogenic shock. The management of critically ill cancer patients poses a unique challenge and requires a multidisciplinary approach. Moreover, no etiologic therapy is available, and only supportive measures can be implemented.
Collapse
Affiliation(s)
- Andrea Montisci
- Division of Cardiothoracic Intensive Care, Azienda Socio-Sanitaria Territoriale (ASST) Spedali Civili, Brescia, Italy
| | - Vittorio Palmieri
- Department of Cardiac Surgery and Transplantation, Ospedali dei Colli Monaldi-Cotugno-CTO, Naples, Italy
| | - Jennifer E Liu
- Department of Medicine/Cardiology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Maria T Vietri
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Silvia Cirri
- Department of Anesthesia and Intensive Care, Istituto Clinico Sant'Ambrogio, Milan, Italy
| | | | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, University Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico - Synlab Diagnostica Nucleare (IRCCS SDN), Naples, Italy
| |
Collapse
|
11
|
Schiano C, D'Armiento M, Franzese M, Castaldo R, Saccone G, de Nigris F, Grimaldi V, Soricelli A, D'Armiento FP, Zullo F, Napoli C. DNA Methylation Profile of the SREBF2 Gene in Human Fetal Aortas. J Vasc Res 2021; 59:61-68. [PMID: 34535602 DOI: 10.1159/000518513] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/13/2021] [Indexed: 11/19/2022] Open
Abstract
Increasing evidence suggests that maternal cholesterol represents an important risk factor for atherosclerotic disease in offspring already during pregnancy, although the underlying mechanisms have not yet been elucidated. Eighteen human fetal aorta samples were collected from the spontaneously aborted fetuses of normal cholesterolemic and hypercholesterolemic mothers. Maternal total cholesterol levels were assessed during hospitalization. DNA methylation profiling of the whole SREBF2 gene CpG island was performed (p value <0.05). The Mann-Whitney U test was used for comparison between the 2 groups. For the first time, our study revealed that in fetal aortas obtained from hypercholesterolemic mothers, the SREBF2 gene shows 4 significant differentially hypermethylated sites in the 5'UTR-CpG island. This finding indicates that more effective long-term primary cardiovascular prevention programs need to be designed for the offspring of mothers with hypercholesterolemia. Further studies should be conducted to clarify the epigenetic mechanisms underlying the association between early atherogenesis and maternal hypercholesterolemia during pregnancy.
Collapse
Affiliation(s)
- Concetta Schiano
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli,", Naples, Italy
| | - Maria D'Armiento
- Pathology Unit, Department of Public Health, School of Medicine, University of Naples "Federico II,", Naples, Italy
| | | | | | - Gabriele Saccone
- Gynecology and Obstetrics Unit, Department of Neurosciences, Reproductive Sciences and Dentistry, School of Medicine, University of Naples "Federico II,", Naples, Italy
| | - Filomena de Nigris
- Department of Precision Medicine, University of Campania "L. Vanvitelli,", Naples, Italy
| | | | - Andrea Soricelli
- IRCCS SDN, Naples, Italy.,Department of Exercise and Wellness Sciences, University of Naples "Parthenope,", Naples, Italy
| | - Francesco Paolo D'Armiento
- Pathology Unit, Department of Public Health, School of Medicine, University of Naples "Federico II,", Naples, Italy
| | - Fulvio Zullo
- Gynecology and Obstetrics Unit, Department of Neurosciences, Reproductive Sciences and Dentistry, School of Medicine, University of Naples "Federico II,", Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli,", Naples, Italy.,IRCCS SDN, Naples, Italy.,Clinical Department of Internal Medicine and Specialistic Units, Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology (SIMT), Regional Reference Laboratory of Transplant Immunology (LIT), Azienda Universitaria Policlinico (AOU), Naples, Italy
| |
Collapse
|
12
|
Infante T, Franzese M, Ruocco A, Schiano C, Affinito O, Pane K, Memoli D, Rizzo F, Weisz A, Bontempo P, Grimaldi V, Berrino L, Soricelli A, Mauro C, Napoli C. ABCA1, TCF7, NFATC1, PRKCZ, and PDGFA DNA methylation as potential epigenetic-sensitive targets in acute coronary syndrome via network analysis. Epigenetics 2021; 17:547-563. [PMID: 34151742 DOI: 10.1080/15592294.2021.1939481] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Acute coronary syndrome (ACS) is the most severe clinical manifestation of coronary heart disease.We performed an epigenome-wide analysis of circulating CD4+ and CD8+ T cells isolated from ACS patients and healthy subjects (HS), enrolled in the DIANA clinical trial, by reduced-representation bisulphite sequencing (RRBS). In CD4+ T cells, we identified 61 differentially methylated regions (DMRs) associated with 57 annotated genes (53% hyper- and 47% hypo-methylated) by comparing ACS patients vs HS. In CD8+ T cells, we identified 613 DMRs associated with 569 annotated genes (28% hyper- and 72% hypo-methylated) in ACS patients as compared to HS. In CD4+ vs CD8+ T cells of ACS patients we identified 175 statistically significant DMRs associated with 157 annotated genes (41% hyper- and 59% hypo-methylated). From pathway analyses, we selected six differentially methylated hub genes (NFATC1, TCF7, PDGFA, PRKCB, PRKCZ, ABCA1) and assessed their expression levels by q-RT-PCR. We found an up-regulation of selected genes in ACS patients vs HS (P < 0.001). ABCA1, TCF7, PDGFA, and PRKCZ gene expression was positively associated with CK-MB serum concentrations (r = 0.75, P = 0.03; r = 0.760, P = 0.029; r = 0.72, P = 0.044; r = 0.74, P = 0.035, respectively).This pilot study is the first single-base resolution map of DNA methylome by RRBS in CD4+ and CD8+ T cells and provides specific methylation signatures to clarify the role of aberrant methylation in ACS pathogenesis, thus supporting future research for novel epigenetic-sensitive biomarkers in the prevention and early diagnosis of this pathology.
Collapse
Affiliation(s)
- Teresa Infante
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | - Antonio Ruocco
- Unit of Cardiovascular Diseases and Arrhythmias, "Antonio Cardarelli" Hospital, Naples, Italy
| | - Concetta Schiano
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | | | - Domenico Memoli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, SA, Italy.,Genome Research Center for Health, Campus of Medicine, Baronissi, SA, Italy
| | - Francesca Rizzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, SA, Italy.,Genome Research Center for Health, Campus of Medicine, Baronissi, SA, Italy
| | - Alessandro Weisz
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana," University of Salerno, Baronissi, SA, Italy.,Genome Research Center for Health, Campus of Medicine, Baronissi, SA, Italy
| | - Paola Bontempo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Vincenzo Grimaldi
- IRCCS SDN, Naples, Italy.,U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Andrea Soricelli
- IRCCS SDN, Naples, Italy.,Department of Exercise and Wellness Sciences, University of Naples Parthenope, Naples, Italy
| | - Ciro Mauro
- Unit of Cardiovascular Diseases and Arrhythmias, "Antonio Cardarelli" Hospital, Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy.,IRCCS SDN, Naples, Italy
| |
Collapse
|
13
|
Napoli C, Bontempo P, Palmieri V, Coscioni E, Maiello C, Donatelli F, Benincasa G. Epigenetic Therapies for Heart Failure: Current Insights and Future Potential. Vasc Health Risk Manag 2021; 17:247-254. [PMID: 34079271 PMCID: PMC8164213 DOI: 10.2147/vhrm.s287082] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
Despite the current reductionist approach providing an optimal indication for diagnosis and treatment of patients with heart failure with reduced ejection fraction (HFrEF), there are no standard pharmacological therapies for heart failure with preserved ejection fraction (HFpEF). Although in its infancy in cardiovascular diseases, the epigenetic-based therapy ("epidrugs") is capturing the interest of physician community. In fact, an increasing number of controlled clinical trials is evaluating the putative beneficial effects of: 1) direct epigenetic-oriented drugs, eg, apabetalone, and 2) repurposed drugs with a possible indirect epigenetic interference, eg, metformin, statins, sodium glucose transporter inhibitors 2 (SGLT2i), and omega 3 polyunsaturated fatty acids (PUFAs) in both HFrEF and HFpEF, separately. Apabetalone is the first and unique direct epidrug tested in cardiovascular patients to date, and the BETonMACE trial has reported a reduction in first HF hospitalization (any EF value) and cardiovascular death in patients with type 2 diabetes and recent acute coronary syndrome, suggesting a possible role in secondary prevention. Patients with HFpEF seem to benefit from supplementation to the standard therapy with statins, metformin, and SGLT2i owing to their ability in reducing mortality. In contrast, the vasodilator hydralazine, with or without isosorbide dinitrate, did not provide beneficial effects. In HFrEF, metformin and SGLT2i could reduce the risk of incident HF and mortality in affected patients whereas clinical trials based on statins provided mixed results. Furthermore, PUFAs diet supplementation was significantly associated with reduced cardiovascular risk in both HFpEF and HFrEF. Future large trials will reveal whether direct and indirect epitherapy will remain a work in progress or become a useful way to customize the therapy in the real-world management of HFpEF and HFrEF. Our goal is to discuss the recent advancement in the epitherapy as a possible way to improve personalized therapy of HF.
Collapse
Affiliation(s)
- Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, 80138, Italy
| | - Paola Bontempo
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Naples, 80138, Italy
| | - Vittorio Palmieri
- Department of Cardiac Surgery and Transplantation, Heart Transplantation Unit in Adults of the 'Ospedali dei Colli Monaldi-Cotugno-CTO', Naples, Italy
| | - Enrico Coscioni
- Department of Cardiac Surgery, Azienda Ospedaliera Universitaria San Giovanni di Dio e Ruggi d'Aragona, Salerno, Italy
| | - Ciro Maiello
- Department of Cardiovascular Surgery and Transplants, Monaldi Hospital, Azienda dei Colli, Naples, Italy
| | - Francesco Donatelli
- Chair of Cardiac Surgery, Department of Cardiothoracic Center, Istituto Clinico Sant'Ambrogio, University of Milan, Milan, Italy
| | - Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, 80138, Italy
| |
Collapse
|
14
|
Pasyukova EG, Symonenko AV, Rybina OY, Vaiserman AM. Epigenetic enzymes: A role in aging and prospects for pharmacological targeting. Ageing Res Rev 2021; 67:101312. [PMID: 33657446 DOI: 10.1016/j.arr.2021.101312] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/05/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
The development of interventions aimed at improving healthspan is one of the priority tasks for the academic and public health authorities. It is also the main objective of a novel branch in biogerontological research, geroscience. According to the geroscience concept, targeting aging is an effective way to combat age-related disorders. Since aging is an exceptionally complex process, system-oriented integrated approaches seem most appropriate for such an interventional strategy. Given the high plasticity and adaptability of the epigenome, epigenome-targeted interventions appear highly promising in geroscience research. Pharmaceuticals targeted at mechanisms involved in epigenetic control of gene activity are actively developed and implemented to prevent and treat various aging-related conditions such as cardiometabolic, neurodegenerative, inflammatory disorders, and cancer. In this review, we describe the roles of epigenetic mechanisms in aging; characterize enzymes contributing to the regulation of epigenetic processes; particularly focus on epigenetic drugs, such as inhibitors of DNA methyltransferases and histone deacetylases that may potentially affect aging-associated diseases and longevity; and discuss possible caveats associated with the use of epigenetic drugs.
Collapse
Affiliation(s)
- Elena G Pasyukova
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Kurchatov Sq. 2, Moscow, 123182, Russia
| | - Alexander V Symonenko
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Kurchatov Sq. 2, Moscow, 123182, Russia
| | - Olga Y Rybina
- Institute of Molecular Genetics of National Research Centre "Kurchatov Institute", Kurchatov Sq. 2, Moscow, 123182, Russia; Federal State Budgetary Educational Institution of Higher Education «Moscow Pedagogical State University», M. Pirogovskaya Str. 1/1, Moscow, 119991, Russia
| | | |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW Cardiovascular diseases (CVDs) are typically caused by multifactorial events including mutations in a large number of genes. Epigenetic-derived modifications in the cells are normal but can be amended by aging, lifestyle, and exposure to toxic substances. Major epigenetic modifications are DNA methylation, histone modification, chromatin remodeling as well as the noncoding RNAs. These pivotal players are involved in the epigenetic-induced modifications observed during CVDs. Nevertheless, despite impressive efforts capitalized in epigenetic research in the last 50 years, clinical applications are still not satisfactory. RECENT FINDINGS Briefly, we present some of the recent steps forward in the epigenetic studies of CVDs. There is an increased appreciation for the contribution of epigenetic alterations in the development of CVDs. Now, we have novel epigenetic biomarkers and therapeutic trials with the use of statins, metformin, and some compounds affecting epigenetic pathways including a BET inhibitor apabetalone. The new knowledge of epigenetic regulation is also discussed in the light of precision medicine of CVDs. SUMMARY Epigenetic studies of CVDs have the promise to yield both mechanistic insights as well as adjunct treatments (repurposed drugs and apabetalone). The overall concept of precision medicine is not widely recognized in routine medical practice and the so-called reductionist approach remains the most used way to treat CVD patients.
Collapse
|
16
|
Sarno F, Benincasa G, List M, Barabasi AL, Baumbach J, Ciardiello F, Filetti S, Glass K, Loscalzo J, Marchese C, Maron BA, Paci P, Parini P, Petrillo E, Silverman EK, Verrienti A, Altucci L, Napoli C. Clinical epigenetics settings for cancer and cardiovascular diseases: real-life applications of network medicine at the bedside. Clin Epigenetics 2021; 13:66. [PMID: 33785068 PMCID: PMC8010949 DOI: 10.1186/s13148-021-01047-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
Despite impressive efforts invested in epigenetic research in the last 50 years, clinical applications are still lacking. Only a few university hospital centers currently use epigenetic biomarkers at the bedside. Moreover, the overall concept of precision medicine is not widely recognized in routine medical practice and the reductionist approach remains predominant in treating patients affected by major diseases such as cancer and cardiovascular diseases. By its' very nature, epigenetics is integrative of genetic networks. The study of epigenetic biomarkers has led to the identification of numerous drugs with an increasingly significant role in clinical therapy especially of cancer patients. Here, we provide an overview of clinical epigenetics within the context of network analysis. We illustrate achievements to date and discuss how we can move from traditional medicine into the era of network medicine (NM), where pathway-informed molecular diagnostics will allow treatment selection following the paradigm of precision medicine.
Collapse
Affiliation(s)
- Federica Sarno
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | - Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Albert-Lazlo Barabasi
- Network Science Institute and Department of Physics, Northeastern University, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Network and Data Science, Central European University, Budapest, Hungary
| | - Jan Baumbach
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
- Chair of Computational Systems Biology, University of Hamburg, Notkestrasse 9, Hamburg, Germany
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy
| | | | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Bradley A Maron
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Paola Paci
- Department of Computer, Control, and Management Engineering, Sapienza University, Rome, Italy
| | - Paolo Parini
- Department of Laboratory Medicine and Department of Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Enrico Petrillo
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Antonella Verrienti
- Department of Translational and Precision Medicine, Sapienza University, Rome, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Napoli, Italy.
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", Naples, Italy
- Clinical Department of Internal Medicine and Specialistic Units, AOU, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
17
|
Liu H, Wingert A, Wang J, Zhang J, Wang X, Sun J, Chen F, Khalid SG, Jiang J, Zheng D. Extraction of Coronary Atherosclerotic Plaques From Computed Tomography Imaging: A Review of Recent Methods. Front Cardiovasc Med 2021; 8:597568. [PMID: 33644127 PMCID: PMC7903898 DOI: 10.3389/fcvm.2021.597568] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 01/18/2021] [Indexed: 12/21/2022] Open
Abstract
Background: Atherosclerotic plaques are the major cause of coronary artery disease (CAD). Currently, computed tomography (CT) is the most commonly applied imaging technique in the diagnosis of CAD. However, the accurate extraction of coronary plaque geometry from CT images is still challenging. Summary of Review: In this review, we focused on the methods in recent studies on the CT-based coronary plaque extraction. According to the dimension of plaque extraction method, the studies were categorized into two-dimensional (2D) and three-dimensional (3D) ones. In each category, the studies were analyzed in terms of data, methods, and evaluation. We summarized the merits and limitations of current methods, as well as the future directions for efficient and accurate extraction of coronary plaques using CT imaging. Conclusion: The methodological innovations are important for more accurate CT-based assessment of coronary plaques in clinical applications. The large-scale studies, de-blooming algorithms, more standardized datasets, and more detailed classification of non-calcified plaques could improve the accuracy of coronary plaque extraction from CT images. More multidimensional geometric parameters can be derived from the 3D geometry of coronary plaques. Additionally, machine learning and automatic 3D reconstruction could improve the efficiency of coronary plaque extraction in future studies.
Collapse
Affiliation(s)
- Haipeng Liu
- Research Centre for Intelligent Healthcare, Coventry University, Coventry, United Kingdom.,Faculty of Health, Education, Medicine, and Social Care, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Aleksandra Wingert
- Faculty of Health, Education, Medicine, and Social Care, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Jian'an Wang
- Department of Cardiology, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jucheng Zhang
- Department of Clinical Engineering, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Xinhong Wang
- Department of Radiology, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Jianzhong Sun
- Department of Radiology, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Fei Chen
- Department of Electrical and Electronic Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Syed Ghufran Khalid
- Research Centre for Intelligent Healthcare, Coventry University, Coventry, United Kingdom
| | - Jun Jiang
- Department of Cardiology, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Dingchang Zheng
- Research Centre for Intelligent Healthcare, Coventry University, Coventry, United Kingdom
| |
Collapse
|
18
|
Napoli C, Benincasa G, Schiano C, Salvatore M. Differential epigenetic factors in the prediction of cardiovascular risk in diabetic patients. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2020; 6:239-247. [PMID: 31665258 PMCID: PMC7363021 DOI: 10.1093/ehjcvp/pvz062] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/02/2019] [Accepted: 10/24/2019] [Indexed: 12/30/2022]
Abstract
Hyperglycaemia can strongly alter the epigenetic signatures in many types of human vascular cells providing persistent perturbations of protein–protein interactions both in micro- and macro-domains. The establishment of these epigenetic changes may precede cardiovascular (CV) complications and help us to predict vascular lesions in diabetic patients. Importantly, these epigenetic marks may be transmitted across several generations (transgenerational effect) and increase the individual risk of disease. Aberrant DNA methylation and imbalance of histone modifications, mainly acetylation and methylation of H3, represent key determinants of vascular lesions and, thus, putative useful biomarkers for prevention and diagnosis of CV risk in diabetics. Moreover, a differential expression of some micro-RNAs (miRNAs), mainly miR-126, may be a useful prognostic biomarker for atherosclerosis development in asymptomatic subjects. Recently, also environmental-induced chemical perturbations in mRNA (epitranscriptome), mainly the N6-methyladenosine, have been associated with obesity and diabetes. Importantly, reversal of epigenetic changes by modulation of lifestyle and use of metformin, statins, fenofibrate, and apabetalone may offer useful therapeutic options to prevent or delay CV events in diabetics increasing the opportunity for personalized therapy. Network medicine is a promising molecular-bioinformatic approach to identify the signalling pathways underlying the pathogenesis of CV lesions in diabetic patients. Moreover, machine learning tools combined with tomography are advancing the individualized assessment of CV risk in these patients. We remark the need for combining epigenetics and advanced bioinformatic platforms to improve the prediction of vascular lesions in diabetics increasing the opportunity for CV precision medicine.
Collapse
Affiliation(s)
- Claudio Napoli
- Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Pz. Miraglia 2, Naples 80138, Italy.,IRCCS SDN, via E. Gianturco 113, Naples 80143, Italy
| | - Giuditta Benincasa
- Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Pz. Miraglia 2, Naples 80138, Italy
| | - Concetta Schiano
- Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Pz. Miraglia 2, Naples 80138, Italy
| | | |
Collapse
|
19
|
Benincasa G, Franzese M, Schiano C, Marfella R, Miceli M, Infante T, Sardu C, Zanfardino M, Affinito O, Mansueto G, Sommese L, Nicoletti GF, Salvatore M, Paolisso G, Napoli C. DNA methylation profiling of CD04 +/CD08 + T cells reveals pathogenic mechanisms in increasing hyperglycemia: PIRAMIDE pilot study. Ann Med Surg (Lond) 2020; 60:218-226. [PMID: 33194177 PMCID: PMC7645316 DOI: 10.1016/j.amsu.2020.10.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/08/2020] [Indexed: 12/11/2022] Open
Abstract
Background DNA methylation can play a pathogenic role in the early stages of hyperglycemia linking homeostasis imbalance and vascular damage. Material and methods We investigated DNA methylome by RRBS in CD04+ and CD08+ T cells from healthy subjects (HS) to pre-diabetics (Pre-Diab) and type 2 diabetic (T2D) patients to identify early biomarkers of glucose impairment and vascular damage. Our cross-sectional study enrolled 14 individuals from HS state to increasing hyperglycemia (pilot study, PIRAMIDE trial, NCT03792607). Results Globally, differentially methylated regions (DMRs) were mostly annotated to promoter regions. Hypermethylated DMRs were greater than hypomethylated in CD04+ T cells whereas CD08+ T showed an opposite trend. Moreover, DMRs overlapping between Pre-Diab and T2D patients were mostly hypermethylated in both T cells. Interestingly, SPARC was the most hypomethylated gene in Pre-Diab and its methylation level gradually decreased in T2D patients. Besides, SPARC showed a significant positive correlation with DBP (+0.76), HDL (+0.54), Creatinine (+0.83), LVDd (+0.98), LVSD (+0.98), LAD (+0.98), LVPWd (+0.84), AODd (+0.81), HR (+0.72), Triglycerides (+0.83), LAD (+0.69) and AODd (+0.52) whereas a negative correlation with Cholesterol (−0.52) and LDL (−0.71) in T2D. Conclusion SPARC hypomethylation in CD08+ T cells may be a useful biomarker of vascular complications in Pre-Diab with a possible role for primary prevention warranting further multicenter clinical trials to validate our findings. We conducted the first methylome analysis by RRBS platform in circulating CD04+ and CD08+ T cells in increasing hyperglycemia. This approach has revealed possible biomarkers for cardiovascular and kidney complications in prediabetes. SPARC hypomethylation may underly a pro-fibrotic endophenotype to be validated in larger multicenter trials.
Collapse
Affiliation(s)
- Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | | | - Concetta Schiano
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Raffaele Marfella
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | | | - Teresa Infante
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Celestino Sardu
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | | | | | - Gelsomina Mansueto
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Linda Sommese
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, Plastic Surgery Unit University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | | | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy.,IRCCS SDN, 80143, Naples, Italy.,Clinical Department of Internal Medicine and Specialistics, Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| |
Collapse
|
20
|
Schiano C, Benincasa G, Infante T, Franzese M, Castaldo R, Fiorito C, Mansueto G, Grimaldi V, Della Valle G, Fatone G, Soricelli A, Nicoletti GF, Ruocco A, Mauro C, Salvatore M, Napoli C. Integrated analysis of DNA methylation profile of HLA-G gene and imaging in coronary heart disease: Pilot study. PLoS One 2020; 15:e0236951. [PMID: 32790754 PMCID: PMC7425923 DOI: 10.1371/journal.pone.0236951] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023] Open
Abstract
Aims Immune endothelial inflammation, underlying coronary heart disease (CHD) related phenotypes, could provide new insight into the pathobiology of the disease. We investigated DNA methylation level of the unique CpG island of HLA-G gene in CHD patients and evaluated the correlation with cardiac computed tomography angiography (CCTA) features. Methods Thirty-two patients that underwent CCTA for suspected CHD were enrolled for this study. Obstructive CHD group included fourteen patients, in which there was a stenosis greater than or equal to 50% in one or more of the major coronary arteries detected; whereas subjects with Calcium (Ca) Score = 0, uninjured coronaries and with no obstructive CHD (no critical stenosis, NCS) were considered as control subjects (n = 18). For both groups, DNA methylation profile of the whole 5’UTR-CpG island of HLA-G was measured. The plasma soluble HLA-G (sHLA-G) levels were detected in all subjects by specific ELISA assay. Statistical analysis was performed using R software. Results For the first time, our study reported that 1) a significant hypomethylation characterized three specific fragments (B, C and F) of the 5’UTR-CpG island (p = 0.05) of HLA-G gene in CHD patients compared to control group; 2) the hypomethylation level of one specific fragment of 161bp (+616/+777) positively correlated with coronary Ca score, a relevant parameter of CCTA (p<0.05) between two groups evaluated and was predictive for disease severity. Conclusions Reduced levels of circulating HLA-G molecules could derive from epigenetic marks. Epigenetics phenomena induce hypomethylation of specific regions into 5'UTR-CpG island of HLA-G gene in CHD patients with obstructive non critical stenosis vs coronary stenosis individuals.
Collapse
Affiliation(s)
- Concetta Schiano
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania “L. Vanvitelli”, Naples, Italy
- * E-mail:
| | - Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania “L. Vanvitelli”, Naples, Italy
| | - Teresa Infante
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania “L. Vanvitelli”, Naples, Italy
| | | | | | - Carmela Fiorito
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], University of Campania "L. Vanvitelli", Naples, Italy
| | - Gelsomina Mansueto
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania “L. Vanvitelli”, Naples, Italy
| | - Vincenzo Grimaldi
- Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology [SIMT], Clinical Department of Internal Medicine and Specialistic Units, Regional Reference Laboratory of Transplant Immunology [LIT], Azienda Universitaria Policlinico (AOU), Naples, Italy
| | - Giovanni Della Valle
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Napoli, Italy
| | - Gerardo Fatone
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, Napoli, Italy
| | - Andrea Soricelli
- IRCCSSDN, Naples, Italy
- Department of Exercise and Wellness Sciences, University of Naples Parthenope, Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, Plastic Surgery Unit, University of Campania “L. Vanvitelli”, Naples, Italy
| | - Antonio Ruocco
- Cardiovascular Diseases Department, “Cardarelli Hospital”, Naples, Italy
| | - Ciro Mauro
- Cardiovascular Diseases Department, “Cardarelli Hospital”, Naples, Italy
| | | | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania “L. Vanvitelli”, Naples, Italy
- IRCCSSDN, Naples, Italy
| |
Collapse
|
21
|
Infante T, Del Viscovo L, De Rimini ML, Padula S, Caso P, Napoli C. Network Medicine: A Clinical Approach for Precision Medicine and Personalized Therapy in Coronary Heart Disease. J Atheroscler Thromb 2020; 27:279-302. [PMID: 31723086 PMCID: PMC7192819 DOI: 10.5551/jat.52407] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/24/2019] [Indexed: 12/13/2022] Open
Abstract
Early identification of coronary atherosclerotic pathogenic mechanisms is useful for predicting the risk of coronary heart disease (CHD) and future cardiac events. Epigenome changes may clarify a significant fraction of this "missing hereditability", thus offering novel potential biomarkers for prevention and care of CHD. The rapidly growing disciplines of systems biology and network science are now poised to meet the fields of precision medicine and personalized therapy. Network medicine integrates standard clinical recording and non-invasive, advanced cardiac imaging tools with epigenetics into deep learning for in-depth CHD molecular phenotyping. This approach could potentially explore developing novel drugs from natural compounds (i.e. polyphenols, folic acid) and repurposing current drugs, such as statins and metformin. Several clinical trials have exploited epigenetic tags and epigenetic sensitive drugs both in primary and secondary prevention. Due to their stability in plasma and easiness of detection, many ongoing clinical trials are focused on the evaluation of circulating miRNAs (e.g. miR-8059 and miR-320a) in blood, in association with imaging parameters such as coronary calcifications and stenosis degree detected by coronary computed tomography angiography (CCTA), or functional parameters provided by FFR/CT and PET/CT. Although epigenetic modifications have also been prioritized through network based approaches, the whole set of molecular interactions (interactome) in CHD is still under investigation for primary prevention strategies.
Collapse
Affiliation(s)
- Teresa Infante
- Department of Advanced Clinical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Luca Del Viscovo
- Department of Precision Medicine, Section of Diagnostic Imaging, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | | | - Sergio Padula
- Department of Cardiology, A.O.R.N. Dei Colli, Monaldi Hospital, Naples, Italy
| | - Pio Caso
- Department of Cardiology, A.O.R.N. Dei Colli, Monaldi Hospital, Naples, Italy
| | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
- IRCCS SDN, Naples, Italy
| |
Collapse
|
22
|
Salameh Y, Bejaoui Y, El Hajj N. DNA Methylation Biomarkers in Aging and Age-Related Diseases. Front Genet 2020; 11:171. [PMID: 32211026 PMCID: PMC7076122 DOI: 10.3389/fgene.2020.00171] [Citation(s) in RCA: 168] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 02/13/2020] [Indexed: 12/11/2022] Open
Abstract
Recent research efforts provided compelling evidence of genome-wide DNA methylation alterations in aging and age-related disease. It is currently well established that DNA methylation biomarkers can determine biological age of any tissue across the entire human lifespan, even during development. There is growing evidence suggesting epigenetic age acceleration to be strongly linked to common diseases or occurring in response to various environmental factors. DNA methylation based clocks are proposed as biomarkers of early disease risk as well as predictors of life expectancy and mortality. In this review, we will summarize key advances in epigenetic clocks and their potential application in precision health. We will also provide an overview of progresses in epigenetic biomarker discovery in Alzheimer's, type 2 diabetes, and cardiovascular disease. Furthermore, we will highlight the importance of prospective study designs to identify and confirm epigenetic biomarkers of disease.
Collapse
Affiliation(s)
| | | | - Nady El Hajj
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
23
|
Schiano C, Benincasa G, Franzese M, Della Mura N, Pane K, Salvatore M, Napoli C. Epigenetic-sensitive pathways in personalized therapy of major cardiovascular diseases. Pharmacol Ther 2020; 210:107514. [PMID: 32105674 DOI: 10.1016/j.pharmthera.2020.107514] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The complex pathobiology underlying cardiovascular diseases (CVDs) has yet to be explained. Aberrant epigenetic changes may result from alterations in enzymatic activities, which are responsible for putting in and/or out the covalent groups, altering the epigenome and then modulating gene expression. The identification of novel individual epigenetic-sensitive trajectories at single cell level might provide additional opportunities to establish predictive, diagnostic and prognostic biomarkers as well as drug targets in CVDs. To date, most of studies investigated DNA methylation mechanism and miRNA regulation as epigenetics marks. During atherogenesis, big epigenetic changes in DNA methylation and different ncRNAs, such as miR-93, miR-340, miR-433, miR-765, CHROME, were identified into endothelial cells, smooth muscle cells, and macrophages. During man development, lipid metabolism, inflammation and homocysteine homeostasis, alter vascular transcriptional mechanism of fundamental genes such as ABCA1, SREBP2, NOS, HIF1. At histone level, increased HDAC9 was associated with matrix metalloproteinase 1 (MMP1) and MMP2 expression in pro-inflammatory macrophages of human carotid plaque other than to have a positive effect on toll like receptor signaling and innate immunity. HDAC9 deficiency promoted inflammation resolution and reverse cholesterol transport, which might block atherosclerosis progression and promote lesion regression. Here, we describe main human epigenetic mechanisms involved in atherosclerosis, coronary heart disease, ischemic stroke, peripheral artery disease; cardiomyopathy and heart failure. Different epigenetics mechanisms are activated, such as regulation by circular RNAs, as MICRA, and epitranscriptomics at RNA level. Moreover, in order to open new frontiers for precision medicine and personalized therapy, we offer a panoramic view on the most innovative bioinformatic tools designed to identify putative genes and molecular networks underlying CVDs in man.
Collapse
Affiliation(s)
- Concetta Schiano
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Giuditta Benincasa
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | | | | | | | | | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, Department of Advanced Clinical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy; IRCCS SDN, Naples, Italy
| |
Collapse
|
24
|
Clemente A, Traghella I, Mazzone A, Sbrana S, Vassalle C. Vascular and valvular calcification biomarkers. Adv Clin Chem 2020; 95:73-103. [PMID: 32122525 DOI: 10.1016/bs.acc.2019.08.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Vascular and valvular calcification constitutes a major health problem with serious clinical consequences. It is important for medical laboratorians to improve their knowledge on this topic and to know which biological markers may have a potential interest and might be useful for diagnosis and for management of ectopic calcifications. This review focuses on the pathophysiological mechanisms of vascular and valvular calcification, with emphasis on the mechanisms that are different for the two types of events, which underscore the need for differentiated healthcare, and explain different response to therapy. Available imaging and scoring tools used to assess both vascular and valvular calcification, together with the more studied and reliable biological markers emerging in this field (e.g., Fetuin A and matrix Gla protein), are discussed. Recently proposed functional assays, measuring the propensity of human serum to calcify, appear promising for vascular calcification assessment and are described. Further advancement through omic technologies and statistical tools is also reported. Clinical chemistry and laboratory medicine practitioners overlook this new era that will engage them in the near future, where a close cooperation of professionals with different competencies, including laboratorists, is required. This innovative approach may truly revolutionize practice of laboratory and of whole medicine attitude, making progression in knowledge of pathways relevant to health, as the complex calcification-related pathways, and adding value to patient care, through a precision medicine strategy.
Collapse
|
25
|
Piepoli MF, Abreu A, Albus C, Ambrosetti M, Brotons C, Catapano AL, Corra U, Cosyns B, Deaton C, Graham I, Hoes A, Lochen ML, Matrone B, Redon J, Sattar N, Smulders Y, Tiberi M. Update on cardiovascular prevention in clinical practice: A position paper of the European Association of Preventive Cardiology of the European Society of Cardiology. Eur J Prev Cardiol 2019; 27:181-205. [PMID: 31826679 DOI: 10.1177/2047487319893035] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
European guidelines on cardiovascular prevention in clinical practice were first published in 1994 and have been regularly updated, most recently in 2016, by the Sixth European Joint Task Force. Given the amount of new information that has become available since then, components from the task force and experts from the European Association of Preventive Cardiology of the European Society of Cardiology were invited to provide a summary and critical review of the most important new studies and evidence since the latest guidelines were published. The structure of the document follows that of the previous document and has six parts: Introduction (epidemiology and cost effectiveness); Cardiovascular risk; How to intervene at the population level; How to intervene at the individual level; Disease-specific interventions; and Settings: where to intervene? In fact, in keeping with the guidelines, greater emphasis has been put on a population-based approach and on disease-specific interventions, avoiding re-interpretation of information already and previously considered. Finally, the presence of several gaps in the knowledge is highlighted.
Collapse
Affiliation(s)
- Massimo F Piepoli
- Department of Cardiology, Polichirurgico Hospital G Da Saliceto, Italy.,Institute of Life Sciences, Sant'Anna School of Advanced Studies, Italy
| | - Ana Abreu
- Serviço de Cardiologia, Universidade de Lisboa, Portugal
| | - Christian Albus
- Department of Pshychosomatics and Psychotherapy, University of Cologne, Germany
| | - Marco Ambrosetti
- Department of Cardiac Rehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Institute of Pavia, Italy
| | - Carlos Brotons
- Biomedical Research Institute Sant Pau Research Unit, Sardenya Primary Health Care Center, Spain
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli studi di Milano, Italy
| | - Ugo Corra
- Department of Cardiac Rehabilitation, Istituti Clinici Scientifici Maugeri IRCCS, Institute of Veruno, Italy
| | - Bernard Cosyns
- Department of Cardiology, Universitair Ziekenhuis Brussel, Belgium
| | - Christi Deaton
- Department of Public Health and Primary Care, University of Cambridge, UK
| | - Ian Graham
- Cardiovascular Medicine, Trinity College Dublin, Ireland
| | - Arno Hoes
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Maja-Lisa Lochen
- Department of Community Medicine, UiT the Artic University of Norway, Norway
| | - Benedetta Matrone
- Department of Cardiology, Polichirurgico Hospital G Da Saliceto, Italy
| | - Josep Redon
- INCLIVA Research Institute, University of Valencia, Spain
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, UK
| | - Yvo Smulders
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | | |
Collapse
|
26
|
Benincasa G, Mansueto G, Napoli C. Fluid-based assays and precision medicine of cardiovascular diseases: the ‘hope’ for Pandora’s box? J Clin Pathol 2019; 72:785-799. [DOI: 10.1136/jclinpath-2019-206178] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 12/25/2022]
Abstract
Progresses in liquid-based assays may provide novel useful non-invasive indicators of cardiovascular (CV) diseases. By analysing circulating cells or their products in blood, saliva and urine samples, we can investigate molecular changes present at specific time points in each patient allowing sequential monitoring of disease evolution. For example, an increased number of circulating endothelial cells may be a diagnostic biomarker for diabetic nephropathy and heart failure with preserved ejection fraction. The assessment of circulating cell-free DNA (cfDNA) levels may be useful to predict severity of acute myocardial infarction, as well as diagnose heart graft rejection. Remarkably, circulating epigenetic biomarkers, including DNA methylation, histone modifications and non-coding RNAs are key pathogenic determinants of CV diseases representing putative useful biomarkers and drug targets. For example, the unmethylated FAM101A gene may specifically trace cfDNA derived from cardiomyocyte death providing a powerful diagnostic biomarker of apoptosis during ischaemia. Moreover, changes in plasma levels of circulating miR-92 may predict acute coronary syndrome onset in patients with diabetes. Now, network medicine provides a framework to analyse a huge amount of big data by describing a CV disease as a result of a chain of molecular perturbations rather than a single defect (reductionism). We outline advantages and challenges of liquid biopsy with respect to traditional tissue biopsy and summarise the main completed and ongoing clinical trials in CV diseases. Furthermore, we discuss the importance of combining fluid-based assays, big data and network medicine to improve precision medicine and personalised therapy in this field.
Collapse
|
27
|
Correlation of Circulating miR-765, miR-93-5p, and miR-433-3p to Obstructive Coronary Heart Disease Evaluated by Cardiac Computed Tomography. Am J Cardiol 2019; 124:176-182. [PMID: 31084998 DOI: 10.1016/j.amjcard.2019.04.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/01/2019] [Accepted: 04/05/2019] [Indexed: 12/13/2022]
Abstract
Epigenetic-sensitive mechanisms may be correlated both to pathogenesis and prognosis of coronary heart disease (CHD). We prospectively investigated some plasma circulating microRNA levels in patients undergoing cardiac computed tomography for suspected CHD (n = 95). We show that let-7c-5p, miR-765, miR-483-5p, miR-31-5p, and miR-206 were upregulated in CHD patients (n = 66) versus healthy subjects HS (n = 29); moreover, let-7c-5p, miR-765, miR- 483-5p showed higher expression in obstructive CHD (n = 36) compared to no obstructive CHD patients (n = 66). Remarkably, miR-765, miR-93-5p, and miR-433-3p showed an upregulation in patients with critical coronary stenosis. Multivariate regression analysis demonstrated that miR-765, miR-31-5p, and miR-206 were independently associated with CHD while circulating levels of miR-765 (p = 0.035), miR-433-3p (p = 0.043), and miR-93-5p (p = 0.041) were significantly higher in critical stenosis patients. Receiver operating characteristic curve analysis revealed a good performance for miR-765, miR-93-5p, and miR-433-3p on predicting CHD severity. In conclusion, our study represents a combined epigenetic/imaging approach useful to support the diagnosis and prediction of CHD.
Collapse
|