1
|
Pietruszyńska-Reszetarska A, Pietruszyński R, Irzmański R. The Significance of Genetically Determined Methylation and Folate Metabolism Disorders in the Pathogenesis of Coronary Artery Disease: A Target for New Therapies? Int J Mol Sci 2024; 25:6924. [PMID: 39000032 PMCID: PMC11241586 DOI: 10.3390/ijms25136924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Methylation is a biochemical process involving the addition of a methyl group (-CH3) to various chemical compounds. It plays a crucial role in maintaining the homeostasis of the endothelium, which lines the interior surface of blood vessels, and has been linked, among other conditions, to coronary artery disease (CAD). Despite significant progress in CAD diagnosis and treatment, intensive research continues into genotypic and phenotypic CAD biomarkers. This review explores the significance of the methylation pathway and folate metabolism in CAD pathogenesis, with a focus on endothelial dysfunction resulting from deficiency in the active form of folate (5-MTHF). We discuss emerging areas of research into CAD biomarkers and factors influencing the methylation process. By highlighting genetically determined methylation disorders, particularly the MTHFR polymorphism, we propose the potential use of the active form of folate (5-MTHF) as a novel CAD biomarker and personalized pharmaceutical for selected patient groups. Our aim is to improve the identification of individuals at high risk of CAD and enhance their prognosis.
Collapse
Affiliation(s)
| | - Robert Pietruszyński
- Cardiology Outpatient Clinic, Military Medical Academy Memorial Teaching Hospital of the Medical University of Lodz—Central Veterans’ Hospital, 90-549 Lodz, Poland;
| | - Robert Irzmański
- Department of Internal Medicine, Rehabilitation and Physical Medicine, Medical University of Lodz, 90-645 Lodz, Poland;
| |
Collapse
|
2
|
Gaber MA, Omar OHM, Meki ARMA, Nassar AY, Hassan AKM, Mahmoud MS. The significance of PCSK-9's level and polymorphism in premature coronary artery disease: Relation to risk and severity. Clin Biochem 2024; 125:110729. [PMID: 38342398 DOI: 10.1016/j.clinbiochem.2024.110729] [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: 10/19/2023] [Revised: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK-9) is a circulating protein that plays an important role in lipid metabolism and is linked to inflammation, which has implications for atherosclerosis and its severe cardiac effects. We studied the potential association of the PCSK-9 gene single nucleotide polymorphism (SNP), Oxidized low-density lipoprotein receptor 1- (OLR-1), and caspase-3 serum levels with the risk and severity of premature coronary artery disease (PCAD). The potential contribution of PCSK-9 serum level to the severity of PCAD patients was also assessed. METHOD This case-control study included 120 PCAD patients (age < 45), and 60 age matched healthy controls. Serum PCSK-9 and caspase-3 levels and clinical characteristics were recorded. SYNTAX score was calculated to estimate the severity of the coronary artery lesions. The SNP rs2483205 of the PCSK-9 gene and the rs11053646 of the OLR-1gene were genotyped in all participants. RESULTS Serum PCSK-9 levels were higher in PCAD patients and were significantly different among the three SYNTAX score groups (SS ≤ 12, 12 < SS ≤ 21.5, and SS > 21.5). The diagnostic cutoff values of PCSK-9 and caspase-3 levels for PCAD were > 3.2 ng/mL for both, yielding an area under the curve (AUC) of 0.98 and 0.92, sensitivity of 85 %, 98 %, and specificity of 99.5 %, 93 % for PCSK-9 and caspase-3, respectively. The genotypes TT + CT vs. CC of PCSK-9's rs2483205 SNP presented a higher risk for PCAD and higher SYNTAX scores. Furthermore, the rs11053646 SNP of OLR-1 presented the CG genotype as more risky and having higher SYNTAX scores. CONCLUSION Circulating PCSK9 and caspase-3 concentrations were higher in PCAD patients and were associated with CAD severity. The SNPs of PCSK-9 (rs2483205) and OLR-1 (rs11053646) were associated with PCAD and its severity.
Collapse
Affiliation(s)
- Marwa A Gaber
- Medical Biochemistry Department, Faculty of Medicine, Assiut University, Assiut, Egypt.
| | - Omnia H M Omar
- Assiut International Center of Nanomedicine, Al-rajhy Liver Hospital, Assiut University, Assiut, Egypt
| | - Abdel-Raheim M A Meki
- Medical Biochemistry Department, Faculty of Medicine, Assiut University, Assiut, Egypt; Biochemistry Department, Faculty of Pharmacy, Sphinx University, New Assiut, Egypt
| | - Ahmed Y Nassar
- Medical Biochemistry Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Ayman K M Hassan
- Cardiology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Marwan S Mahmoud
- Cardiology Department, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
3
|
Awan Z, Batran A, Al-Allaf FA, Alharbi RS, Hegazy GA, Jamalalail B, Almansouri M, Bima AI, Almukadi H, Kutbi HI, Altayar AE, Banaganapalli B, Shaik NA. Identification and functional characterization of two rare LDLR stop gain variants (p.C231* and p.R744*) in Saudi familial hypercholesterolemia patients. Panminerva Med 2023; 65:479-490. [PMID: 35274909 DOI: 10.23736/s0031-0808.22.04612-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is a globally underdiagnosed inherited metabolic disorder. Owing to limited published data from Arab world, this study was conducted with the aim of identifying the genetic and molecular basis of FH in highly consanguineous Saudi population. METHODS We performed clinical screening, biochemical profiling, whole exome sequencing and variant segregation analysis of two Saudi FH families. Additionally, 500 normolipic individuals were screened to ensure the absence of FH variant in general Saudi population. Functional characterization of FH variants on secondary structure characteristics of RNA and protein molecules was performed using different bioinformatics modelling approaches. RESULTS WES analysis identified two independent rare LDLR gene stop gain variants (p.C231* and p.R744*) consistent to the clinical presentation of FH patients from two different families. RNAfold analysis has shown that both variants were predicted to disturb the free energy dynamics of LDLR mRNA molecule and destabilize its folding pattern and function. PSIPRED based structural modelling analysis has suggested that both variants bring drastic changes disturbing the secondary structural elements of LDLR molecule. The p.C231* and p.R744* variants are responsible for partial or no protein product, thus they are class 1 variants causing loss of function (LoF) LDLR variants. CONCLUSIONS This study highlights the effectiveness of the WES, sanger sequencing, and computational analysis in expanding FH variant spectrum in culturally distinct populations like Saudi Arabia. Genetic testing of FH patients is very essential in better clinical diagnosis, screening, treatment, and management and prevention of cardiovascular disease burden in the society.
Collapse
Affiliation(s)
- Zuhier Awan
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alhanuf Batran
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Faisal A Al-Allaf
- Department of Medical Genetics, Faculty of Medicine, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Raneem S Alharbi
- Department of Genetics, Al Borg Medical Laboratories, Jeddah, Saudi Arabia
| | - Gehan A Hegazy
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Bassam Jamalalail
- Department of Genetics, Al Borg Medical Laboratories, Jeddah, Saudi Arabia
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Majid Almansouri
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulhadi I Bima
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Haifa Almukadi
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaiziz University, Jeddah, Saudi Arabia
| | - Hussam I Kutbi
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed E Altayar
- Department of Genetic Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Babajan Banaganapalli
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noor A Shaik
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia -
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
4
|
Dragoș D, Timofte D, Georgescu MT, Manea MM, Vacaroiu IA, Ionescu D, Balcangiu-Stroescu AE. Cardiovascular Calcifications Are Correlated with Inflammation in Hemodialysis Patients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1801. [PMID: 37893519 PMCID: PMC10608311 DOI: 10.3390/medicina59101801] [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: 08/18/2023] [Revised: 10/02/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: The main cause of morbidity and mortality in hemodialysis patients is cardiovascular disease, which is quite common. The main objective of our study was to investigate the relationship between oxidative stress, inflammation, and vascular and valvular calcifications in hemodialysis patients. Materials and Methods: This observational study had 54 hemodialysis patients, with an average age of 60.46 ± 13.18 years. Cardiovascular ultrasound was used to detect and/or measure aortic and mitral valve calcifications, carotid and femoral atheroma plaques, and common carotid intima-media thickness. The aortic calcification score was determined using a lateral abdomen plain radiograph. The inflammatory, oxidative, metabolic, and dietary statuses, as well as demographic characteristics, were identified. Results: There were significant correlations between the levels of IL-6 and carotid plaque number (p = 0.003), fibrinogen level and aortic valve calcifications (p = 0.05), intima-media thickness (p = 0.0007), carotid plaque number (p = 0.035), femoral plaque number (p = 0.00014), and aortic calcifications score (p = 0.0079). Aortic annulus calcifications (p = 0.03) and intima-media thickness (p = 0.038) were adversely linked with TNF-α. Nutrition parameters were negatively correlated with atherosclerosis markers: number of carotid plaques with albumin (p = 0.013), body mass index (p = 0.039), and triglycerides (p = 0.021); number of femoral plaques with phosphorus (0.013), aortic calcifications score with albumin (p = 0.051), intima-media thickness with LDL-cholesterol (p = 0.042). Age and the quantity of carotid plaques, femoral plaques, and aortic calcifications were linked with each other (p = 0.0022, 0.00011, and 0.036, respectively). Aortic annulus calcifications (p = 0.011), aortic valve calcifications (p = 0.023), and mitral valve calcifications (p = 0.018) were all associated with an increased risk of death. Conclusions: Imaging measures of atherosclerosis are adversely connected with dietary status and positively correlated with markers of inflammation and risk of mortality.
Collapse
Affiliation(s)
- Dorin Dragoș
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- 1st Internal Medicine Clinic, University Emergency Hospital Bucharest, Splaiul Independentei nr 169, Sect 5, 050098 Bucharest, Romania
| | - Delia Timofte
- Department of Dialysis, University Emergency Hospital Bucharest, Splaiul Independentei nr 168, Sect 5, 050098 Bucharest, Romania
| | - Mihai-Teodor Georgescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- Department of Radiotherapy, Prof. Dr. Al. Trestioreanu Institute of Oncology Bucharest, Șos. Fundeni nr 252, Sect 2, 022328 Bucharest, Romania
| | - Maria-Mirabela Manea
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- National Institute of Neurology and Cerebrovascular Diseases, Șos. Berceni nr 10–12, Sect 4, 041915 Bucharest, Romania
| | - Ileana Adela Vacaroiu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- Nephrology and Dialysis Clinic, “Sf. Ioan” Emergency Clinical Hospital, Șos. Vitan-Barzești nr 12, 042122 Bucharest, Romania
| | - Dorin Ionescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania (I.A.V.)
- Nephrology Clinic, University Emergency Hospital, Splaiul Independentei nr 169, Sect 5, 050098 Bucharest, Romania
| | - Andra-Elena Balcangiu-Stroescu
- Faculty of Dental Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu nr 37, Sect 2, 020021 Bucharest, Romania
| |
Collapse
|
5
|
Hallak AO, Hallak FZ, Hallak YO, Hallak OO, Hayson AW, Tanami SA, Bennett WL, Lavie CJ. Exercise Therapy in the Management of Peripheral Arterial Disease. Mayo Clin Proc Innov Qual Outcomes 2023; 7:476-489. [PMID: 37823000 PMCID: PMC10562863 DOI: 10.1016/j.mayocpiqo.2023.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023] Open
Abstract
The incidence and prevalence of peripheral artery disease (PAD) are increasing globally and have a marked economic burden in the United States. The American Heart Association/American College of Cardiology guidelines recommend exercise therapy as a Class 1A, but its utilization remains suboptimal. This state-of-the-art review aims to provide a comprehensive review of the most updated information available on PAD, along with its risk factors, management options, outcomes, economic burden, and the role of exercise therapy in managing PAD.
Collapse
Affiliation(s)
- Ahmad O. Hallak
- Department of Cardiology, Ochsner Medical Center, New Orleans, LA
| | | | - Yusuf O. Hallak
- School of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | | | - Aaron W. Hayson
- Department of Vascular Surgery, Ochsner Medical Center, New Orleans, LA
| | - Sadia A. Tanami
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Amarillo
| | | | - Carl J. Lavie
- Department of Cardiology, Ochsner Medical Center, New Orleans, LA
| |
Collapse
|
6
|
Boshev M, Stankovic S, Panov S, Josifovska S, Georgiev A, Poposka L, Pejkov H. Association of the Polymorphism rs3918242 of the Matrix Metalloproteinase-9 Gene with Coronary Artery Disease in a Younger Population. Pril (Makedon Akad Nauk Umet Odd Med Nauki) 2023; 44:31-39. [PMID: 37453108 DOI: 10.2478/prilozi-2023-0022] [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: 07/18/2023]
Abstract
Coronary artery disease (CAD) is a complex disease resulting from the interaction of numerous so-called traditional risk factors and comorbid conditions on the one side (such as dyslipidemia, smoking, obesity, diabetes, hypertension) and genetic factors on the other. The evidence of a genetic contribution to the development of CAD, especially in the last 2 decades is consistent. It is important that a number of established gene polymorphisms in the younger CAD population are in the genes involved in the inflammatory response and tissue maintenance and remodeling processes. The aim of this study is to investigate the association of the rs3918242 polymorphism of the matrix metal-loproteinase 9 (MMP9) gene with the coronary artery disease in the younger population. In this observational genetic-association study of cases and controls, the demographic, clinical, laboratory and genetic data of the younger population in a group of selected 70 CAD patients aged up to 45 years were analyzed, of which 35 patients have negative and 35 have positive coronary angiography finding, and 43 are men and 27 are women. The analysis of the genotypic and allelic frequency determined an association of the polymorphism and the occurrence of the positive coronary angiographic findings in the population of patients under the age of 45. The carriers of the heterozygous genotype CT have almost 5 times higher probability of having a positive coronary angiography finding compared to the carriers of the reference homozygous genotype CC (p=0.012). Thus, this parameter could be used for clinical risk assessment for the development of CAD.
Collapse
Affiliation(s)
- Marjan Boshev
- 1University Clinic of Cardiology, Skopje, RN Macedonia
- 4Medical Faculty, University "Ss. Cyril and Methodius" in Skopje, RN Macedonia
| | - Svetlana Stankovic
- 2University Clinic of Hematology, Skopje, RN Macedonia
- 4Medical Faculty, University "Ss. Cyril and Methodius" in Skopje, RN Macedonia
| | - Sasho Panov
- 3Laboratory for Molecular Biology, Faculty of Natural Sciences and Mathematics, University "Ss. Cyril and Methodius" in Skopje, RN Macedonia
| | - Slavica Josifovska
- 3Laboratory for Molecular Biology, Faculty of Natural Sciences and Mathematics, University "Ss. Cyril and Methodius" in Skopje, RN Macedonia
| | - Antonio Georgiev
- 1University Clinic of Cardiology, Skopje, RN Macedonia
- 4Medical Faculty, University "Ss. Cyril and Methodius" in Skopje, RN Macedonia
| | - Lidija Poposka
- 1University Clinic of Cardiology, Skopje, RN Macedonia
- 4Medical Faculty, University "Ss. Cyril and Methodius" in Skopje, RN Macedonia
| | - Hristo Pejkov
- 1University Clinic of Cardiology, Skopje, RN Macedonia
- 4Medical Faculty, University "Ss. Cyril and Methodius" in Skopje, RN Macedonia
| |
Collapse
|
7
|
Meza-Alvarado JC, Page RA, Mallard B, Bromhead C, Palmer BR. VEGF-A related SNPs: a cardiovascular context. Front Cardiovasc Med 2023; 10:1190513. [PMID: 37288254 PMCID: PMC10242119 DOI: 10.3389/fcvm.2023.1190513] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/27/2023] [Indexed: 06/09/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Currently, cardiovascular disease risk algorithms play a role in primary prevention. However, this is complicated by a lack of powerfully predictive biomarkers that could be observed in individuals before the onset of overt symptoms. A key potential biomarker for heart disease is the vascular endothelial growth factor (VEGF-A), a molecule that plays a pivotal role in blood vessel formation. This molecule has a complex biological role in the cardiovascular system due to the processes it influences, and its production is impacted by various CVD risk factors. Research in different populations has shown single nucleotide polymorphisms (SNPs) may affect circulating VEGF-A plasma levels, with some variants associated with the development of CVDs, as well as CVD risk factors. This minireview aims to give an overview of the VEGF family, and of the SNPs reported to influence VEGF-A levels, cardiovascular disease, and other risk factors used in CVD risk assessments.
Collapse
Affiliation(s)
| | | | | | | | - B. R. Palmer
- School of Health Sciences, Massey University, Wellington, New Zealand
| |
Collapse
|
8
|
Mendonça MI, Pereira A, Monteiro J, Sousa JA, Santos M, Temtem M, Borges S, Henriques E, Rodrigues M, Sousa AC, Ornelas I, Freitas AI, Brehm A, Drumond A, Palma Dos Reis R. Impact of genetic information on coronary disease risk in Madeira: The GENEMACOR study. Rev Port Cardiol 2023; 42:193-204. [PMID: 36265803 DOI: 10.1016/j.repc.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 01/05/2022] [Accepted: 01/18/2022] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Coronary artery disease (CAD), characterized by an atherogenic process in the coronary arteries, is one of the leading causes of death in Madeira. The GENEMACOR (GENEs in MAdeira and CORonary Disease) study sought to investigate the main risk factors - environmental and genetic - and estimate whether a genetic risk score (GRS) improves CAD prediction, discrimination and reclassification. METHODS Traditional risk factors and 33 CAD genetic variants were considered in a case-control study with 3139 individuals (1723 patients and 1416 controls). The multivariate analysis assessed the likelihood of CAD. A multiplicative GRS (mGRS) was created, and two models (with and without mGRS) were prepared. Two areas under receiver operating characteristic curve (area under curve (AUC)) were analyzed and compared to discriminate CAD likelihood. Net reclassification improvement (NRI) and integrated discrimination index (IDI) were used to reclassify the population. RESULTS All traditional risk factors were strong and independent predictors of CAD, with smoking being the most significant (OR 3.25; p<0.0001). LPA rs3798220 showed a higher CAD likelihood (odds ratio 1.45; p<0.0001). Individuals in the fourth mGRS quartile had an increased CAD probability of 136% (p<0.0001). A traditional risk factor-based model estimated an AUC of 0.73, rising to 0.75 after mGRS inclusion (p<0.0001), revealing a better fit. Continuous NRI better reclassified 28.1% of the population, and categorical NRI mainly improved the reclassification of the intermediate risk group. CONCLUSIONS CAD likelihood was influenced by traditional risk factors and genetic variants. Incorporating GRS into the traditional model improved CAD predictive capacity, discrimination and reclassification. These approaches may provide helpful diagnostic and therapeutic advances, especially in the intermediate risk group.
Collapse
Affiliation(s)
- Maria Isabel Mendonça
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal.
| | - Andreia Pereira
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal; Serviço de Cardiologia, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - Joel Monteiro
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal; Serviço de Cardiologia, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - João Adriano Sousa
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal; Serviço de Cardiologia, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - Marina Santos
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal; Serviço de Cardiologia, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - Margarida Temtem
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal; Serviço de Cardiologia, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - Sofia Borges
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - Eva Henriques
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - Mariana Rodrigues
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - Ana Célia Sousa
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - Ilídio Ornelas
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - Ana Isabel Freitas
- Centro de Investigação Dra. Maria Isabel Mendonça, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - António Brehm
- Serviço de Cardiologia, Hospital Dr. Nélio Mendonça, SESARAM EPERAM, Funchal, Portugal
| | - António Drumond
- Laboratório de Genética Humana, Universidade da Madeira, Funchal, Portugal
| | | |
Collapse
|
9
|
Poli A, Catapano AL, Corsini A, Manzato E, Werba JP, Catena G, Cetin I, Cicero AFG, Cignarella A, Colivicchi F, Consoli A, Landi F, Lucarelli M, Manfellotto D, Marrocco W, Parretti D, Perrone Filardi P, Pirillo A, Sesti G, Volpe M, Marangoni F. LDL-cholesterol control in the primary prevention of cardiovascular diseases: An expert opinion for clinicians and health professionals. Nutr Metab Cardiovasc Dis 2023; 33:245-257. [PMID: 36566123 DOI: 10.1016/j.numecd.2022.10.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/04/2022] [Indexed: 11/29/2022]
Abstract
AIMS Although adequate clinical management of patients with hypercholesterolemia without a history of known cardiovascular disease is essential for prevention, these subjects are often disregarded. Furthermore, the scientific literature on primary cardiovascular prevention is not as rich as that on secondary prevention; finally, physicians often lack adequate tools for the effective management of subjects in primary prevention and have to face some unsolved relevant issues. This document aims to discuss and review the evidence available on this topic and provide practical guidance. DATA SYNTHESIS Available algorithms and risk charts represent the main tool for the assessment of cardiovascular risk in patients in primary prevention. The accuracy of such an estimate can be substantially improved considering the potential contribution of some additional risk factors (C-reactive protein, lipoprotein(a), family history of cardiovascular disease) and conditions (environmental pollution, sleep quality, socioeconomic status, educational level) whose impact on the cardiovascular risk has been better understood in recent years. The availability of non-invasive procedures to evaluate subclinical atherosclerosis may help to identify subjects needing an earlier intervention. Unveiling the presence of these conditions will improve cardiovascular risk estimation, granting a more appropriate intervention. CONCLUSIONS The accurate assessment of cardiovascular risk in subjects in primary prevention with the use of algorithms and risk charts together with the evaluation of additional factors will allow physicians to approach each patient with personalized strategies, which should translate into an increased adherence to therapy and, as a consequence, a reduced cardiovascular risk.
Collapse
Affiliation(s)
- Andrea Poli
- NFI - Nutrition Foundation of Italy, Milan, Italy.
| | - Alberico L Catapano
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy; Center for the Study of Dyslipidaemias, IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Enzo Manzato
- Department of Medicine, University of Padova, Padova, Italy; SISA - Italian Society for the Study of Atherosclerosis, Italy
| | - José Pablo Werba
- Unit of Atherosclerosis Prevention, Monzino Cardiology Center, IRCCS, Milan, Italy
| | | | - Irene Cetin
- Department of Woman, Mother and Neonate Hospital Buzzi, Milan, University of Milan, Italy; SIGO - Italian Society of Gynecology and Obstetrics, Italy
| | - Arrigo F G Cicero
- Hypertension and Cardiovascular Risk Research Center, Medical and Surgical Sciences Department, IRCCS AOU di Bologna, Bologna, Italy; SINut - Italian Nutraceutical Society, Italy
| | - Andrea Cignarella
- Department of Medicine, University of Padova, Padova, Italy; Italian Research Center for Gender Health and Medicine, Italy
| | - Furio Colivicchi
- Division of Clinical Cardiology, San Filippo Neri Hospital, Rome, Italy; ANMCO - Italian National Association of Hospital Cardiologists, Italy
| | - Agostino Consoli
- Department of Medicine and Aging Sciences, University G. D'Annunzio, Chieti, Italy; SID - Italian Society of Diabetology, Italy
| | - Francesco Landi
- Fondazione Policlinico Universitario A. Gemelli IRCSS, Rome, Italy; SIGG - Italian Society of Gerontology and Geriatrics, Italy
| | - Maurizio Lucarelli
- SNaMID - National Society of Medical Education in General Practice, Italy
| | - Dario Manfellotto
- Department of Internal Medicine, Fatebenefratelli Hospital, Isola Tiberina, Rome, Italy; FADOI - Federation of Associations of Hospital Internists, Italy
| | - Walter Marrocco
- SIMPeSV and FIMMG - Italian Society of Preventive and Lifestyle Medicine and Italian Federation of General Practitioners, Italy
| | | | - Pasquale Perrone Filardi
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy; SIC - Italian Society of Cardiology, Italy
| | - Angela Pirillo
- Center for the Study of Dyslipidaemias, IRCCS MultiMedica, Sesto S. Giovanni, Milan, Italy; Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, Milan, Italy
| | - Giorgio Sesti
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Rome, Italy; SIMI - Italian Society of Internal Medicine, Italy
| | - Massimo Volpe
- Department of Clinical and Molecular Medicine, Sapienza University of Rome, Italy; SIPREC - Italian Society for Cardiovascular Prevention, Italy
| | | |
Collapse
|
10
|
Tcheandjieu C, Zhu X, Hilliard AT, Clarke SL, Napolioni V, Ma S, Lee KM, Fang H, Chen F, Lu Y, Tsao NL, Raghavan S, Koyama S, Gorman BR, Vujkovic M, Klarin D, Levin MG, Sinnott-Armstrong N, Wojcik GL, Plomondon ME, Maddox TM, Waldo SW, Bick AG, Pyarajan S, Huang J, Song R, Ho YL, Buyske S, Kooperberg C, Haessler J, Loos RJF, Do R, Verbanck M, Chaudhary K, North KE, Avery CL, Graff M, Haiman CA, Le Marchand L, Wilkens LR, Bis JC, Leonard H, Shen B, Lange LA, Giri A, Dikilitas O, Kullo IJ, Stanaway IB, Jarvik GP, Gordon AS, Hebbring S, Namjou B, Kaufman KM, Ito K, Ishigaki K, Kamatani Y, Verma SS, Ritchie MD, Kember RL, Baras A, Lotta LA, Kathiresan S, Hauser ER, Miller DR, Lee JS, Saleheen D, Reaven PD, Cho K, Gaziano JM, Natarajan P, Huffman JE, Voight BF, Rader DJ, Chang KM, Lynch JA, Damrauer SM, Wilson PWF, Tang H, Sun YV, Tsao PS, O'Donnell CJ, Assimes TL. Large-scale genome-wide association study of coronary artery disease in genetically diverse populations. Nat Med 2022; 28:1679-1692. [PMID: 35915156 PMCID: PMC9419655 DOI: 10.1038/s41591-022-01891-3] [Citation(s) in RCA: 123] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/08/2022] [Indexed: 02/03/2023]
Abstract
We report a genome-wide association study (GWAS) of coronary artery disease (CAD) incorporating nearly a quarter of a million cases, in which existing studies are integrated with data from cohorts of white, Black and Hispanic individuals from the Million Veteran Program. We document near equivalent heritability of CAD across multiple ancestral groups, identify 95 novel loci, including nine on the X chromosome, detect eight loci of genome-wide significance in Black and Hispanic individuals, and demonstrate that two common haplotypes at the 9p21 locus are responsible for risk stratification in all populations except those of African origin, in which these haplotypes are virtually absent. Moreover, in the largest GWAS for angiographically derived coronary atherosclerosis performed to date, we find 15 loci of genome-wide significance that robustly overlap with established loci for clinical CAD. Phenome-wide association analyses of novel loci and polygenic risk scores (PRSs) augment signals related to insulin resistance, extend pleiotropic associations of these loci to include smoking and family history, and precisely document the markedly reduced transferability of existing PRSs to Black individuals. Downstream integrative analyses reinforce the critical roles of vascular endothelial, fibroblast, and smooth muscle cells in CAD susceptibility, but also point to a shared biology between atherosclerosis and oncogenesis. This study highlights the value of diverse populations in further characterizing the genetic architecture of CAD.
Collapse
Affiliation(s)
- Catherine Tcheandjieu
- VA Palo Alto Health Care System, Palo Alto, CA, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA.
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA.
| | - Xiang Zhu
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
- Department of Statistics, The Pennsylvania State University, University Park, PA, USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| | | | - Shoa L Clarke
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Valerio Napolioni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Shining Ma
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Kyung Min Lee
- VA Informatics and Computing Infrastructure, VA Salt Lake City Health Care System, Salt Lake City, UT, USA
| | - Huaying Fang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Fei Chen
- Department of Preventive Medicine, Center for Genetic Epidemiology, University of Southern California, Los Angeles, CA, USA
| | - Yingchang Lu
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Noah L Tsao
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sridharan Raghavan
- Medicine Service, VA Eastern Colorado Health Care System, Aurora, CO, USA
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Satoshi Koyama
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Bryan R Gorman
- VA Boston Healthcare System, Boston, MA, USA
- Booz Allen Hamilton, McLean, VA, USA
| | - Marijana Vujkovic
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek Klarin
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- VA Boston Healthcare System, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Vascular Surgery and Endovascular Therapy, University of Florida School of Medicine, Gainesville, FL, USA
- Stanford University School of Medicine, Stanford, CA, USA
| | - Michael G Levin
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Nasa Sinnott-Armstrong
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Genevieve L Wojcik
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Mary E Plomondon
- Department of Medicine, Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- CART Program, VHA Office of Quality and Patient Safety, Washington, DC, USA
| | - Thomas M Maddox
- Healthcare Innovation Lab, JC HealthCare/Washington University School of Medicine, St Louis, MO, USA
- Division of Cardiology, Washington University School of Medicine, St Louis, MO, USA
| | - Stephen W Waldo
- Department of Medicine, Rocky Mountain Regional VA Medical Center, Aurora, CO, USA
- CART Program, VHA Office of Quality and Patient Safety, Washington, DC, USA
- Division of Cardiology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Alexander G Bick
- Department of Biomedical Informatics, Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Saiju Pyarajan
- VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jie Huang
- VA Boston Healthcare System, Boston, MA, USA
- Department of Global Health, Peking University School of Public Health, Beijing, China
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | | | - Yuk-Lam Ho
- VA Boston Healthcare System, Boston, MA, USA
| | - Steven Buyske
- Department of Statistics, Rutgers University, Piscataway, NJ, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ron Do
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marie Verbanck
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- EA 7537 BioSTM, Université de Paris, Paris, France
| | - Kumardeep Chaudhary
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Christy L Avery
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Christopher A Haiman
- Department of Preventive Medicine, Center for Genetic Epidemiology, University of Southern California, Los Angeles, CA, USA
| | - Loïc Le Marchand
- Cancer Epidemiology Program, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Lynne R Wilkens
- Cancer Epidemiology Program, University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, USA
| | - Joshua C Bis
- Department of Medicine, Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Hampton Leonard
- Molecular Genetics Section, Laboratory of Neurogenetics, National Institute on Aging, Bethesda, MD, USA
- Data Tecnica Int'l, LLC, Glen Echo, MD, USA
| | - Botong Shen
- Health Disparities Research Section, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Leslie A Lange
- Department of Medicine, Division of Biomedical Informatics and Personalized Medicine, Aurora, CO, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, Aurora, CO, USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Ayush Giri
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Obstetrics and Gynecology, Division of Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ozan Dikilitas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ian B Stanaway
- Department of Medicine, Division of Nephrology, University of Washington, Seattle, WA, USA
| | - Gail P Jarvik
- Department of Medicine, Medical Genetics, University of Washington School of Medicine, Seattle, WA, USA
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Adam S Gordon
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Scott Hebbring
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI, USA
| | - Bahram Namjou
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kenneth M Kaufman
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kaoru Ito
- Laboratory for Cardiovascular Genomics and Informatics, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Kazuyoshi Ishigaki
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences - The University of Tokyo, Tokyo, Japan
| | - Shefali S Verma
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute for Biomedical Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rachel L Kember
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aris Baras
- Regeneron Genetics Center, Tarrytown, NY, USA
| | | | - Sekar Kathiresan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Verve Therapeutics, Cambridge, MA, USA
| | - Elizabeth R Hauser
- Cooperative Studies Program Epidemiology Center-Durham, Durham VA Health Care System, Durham, NC, USA
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC, USA
| | - Donald R Miller
- Center for Healthcare Organization and Implementation Research, Bedford VA Healthcare System, Bedford, MA, USA
- Center for Population Health, Department of Biomedical and Nutritional Sciences, University of Massachusetts, Lowell, MA, USA
| | - Jennifer S Lee
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Danish Saleheen
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, Division of Cardiology, Columbia University, New York, NY, USA
| | - Peter D Reaven
- Phoenix VA Health Care System, Phoenix, AZ, USA
- College of Medicine, University of Arizona, Phoenix, AZ, USA
| | - Kelly Cho
- VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - J Michael Gaziano
- VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Cardiology Division, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Benjamin F Voight
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Institute of Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel J Rader
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Kyong-Mi Chang
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Julie A Lynch
- VA Salt Lake City Health Care System, Salt Lake City, UT, USA
- College of Nursing and Health Sciences, University of Massachusetts, Boston, MA, USA
| | - Scott M Damrauer
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Peter W F Wilson
- Atlanta VA Medical Center, Atlanta, GA, USA
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hua Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yan V Sun
- Atlanta VA Health Care System, Atlanta, GA, USA
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Philip S Tsao
- VA Palo Alto Health Care System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher J O'Donnell
- VA Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Themistocles L Assimes
- VA Palo Alto Health Care System, Palo Alto, CA, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
11
|
Wang K, Shi X, Zhu Z, Hao X, Chen L, Cheng S, Foo RSY, Wang C. Mendelian randomization analysis of 37 clinical factors and coronary artery disease in East Asian and European populations. Genome Med 2022; 14:63. [PMID: 35698167 PMCID: PMC9195360 DOI: 10.1186/s13073-022-01067-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 06/03/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Coronary artery disease (CAD) remains the leading cause of mortality worldwide despite enormous efforts devoted to its prevention and treatment. While many genetic loci have been identified to associate with CAD, the intermediate causal risk factors and etiology have not been fully understood. This study assesses the causal effects of 37 heritable clinical factors on CAD in East Asian and European populations. METHODS We collected genome-wide association summary statistics of 37 clinical factors from the Biobank Japan (42,793 to 191,764 participants) and the UK Biobank (314,658 to 442,817 participants), paired with summary statistics of CAD from East Asians (29,319 cases and 183,134 controls) and Europeans (91,753 cases and 311,344 controls). These clinical factors covered 12 cardiometabolic traits, 13 hematological indices, 7 hepatological and 3 renal function indices, and 2 serum electrolyte indices. We performed univariable and multivariable Mendelian randomization (MR) analyses in East Asians and Europeans separately, followed by meta-analysis. RESULTS Univariable MR analyses identified reliable causal evidence (P < 0.05/37) of 10 cardiometabolic traits (height, body mass index [BMI], blood pressure, glycemic and lipid traits) and 4 other clinical factors related to red blood cells (red blood cell count [RBC], hemoglobin, hematocrit) and uric acid (UA). Interestingly, while generally consistent, we identified population heterogeneity in the causal effects of BMI and UA, with higher effect sizes in East Asians than those in Europeans. After adjusting for cardiometabolic factors in multivariable MR analysis, red blood cell traits (RBC, meta-analysis odds ratio 1.07 per standard deviation increase, 95% confidence interval 1.02-1.13; hemoglobin, 1.10, 1.03-1.16; hematocrit, 1.10, 1.04-1.17) remained significant (P < 0.05), while UA showed an independent causal effect in East Asians only (1.12, 1.06-1.19, P = 3.26×10-5). CONCLUSIONS We confirmed the causal effects of 10 cardiometabolic traits on CAD and identified causal risk effects of RBC, hemoglobin, hematocrit, and UA independent of traditional cardiometabolic factors. We found no causal effects for 23 clinical factors, despite their reported epidemiological associations. Our findings suggest the physiology of red blood cells and the level of UA as potential intervention targets for the prevention of CAD.
Collapse
Affiliation(s)
- Kai Wang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xian Shi
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ziwei Zhu
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shanshan Cheng
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Roger S Y Foo
- Cardiovascular Research Institute, Centre for Translational Medicine, National University Health System, Singapore, Singapore
- Genome Institute of Singapore, Singapore, Singapore
| | - Chaolong Wang
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Guo J, Gao Y, Ahmed M, Dong P, Gao Y, Gong Z, Liu J, Mao Y, Yue Z, Zheng Q, Li J, Rong J, Zhou Y, An M, Gu L, Zhang J. Serum Homocysteine Level Predictive Capability for Severity of Restenosis Post Percutaneous Coronary Intervention. Front Pharmacol 2022; 13:816059. [PMID: 35685647 PMCID: PMC9171111 DOI: 10.3389/fphar.2022.816059] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Background: In stent restenosis (ISR) is one of the major complications after stent implantation. Thus, there is a growing interest in identifying a biomarker for the onset of ISR. High levels of serum homocysteine (Hcy) have been associated with the progression of cardiovascular disease. Therefore, the study was carried out to quantify the correlation between serum Hcy and ISR severity. Compared with coronary angiography (CAG), Hcy levels provided a significantly better clinical detection of ISR severity after PCI. Methods: A total of 155 patients were recruited from Shanxi Bethune hospital, from 6 months to 2 years post PCI. Serum Hcy levels and postoperative angiography results were used to differentiate the patients into two experimental groups: ISR (>50% diametrical stenosis), and non-ISR. The non-ISR included two subgroups: intimal hyperplasia (10–50% diametrical stenosis), and recovery (<10% diametrical stenosis). In addition, a group of 80 healthy individuals was used as a negative control. The correlation between homocysteine level and ISR severity t was analyzed for all groups. In addition, the correlation between serum Hcy level and the severity of ISR in the experimental group was analyzed by the Pearson correlation test. Results: The serum Hcy level in the experimental group and control group was determined to be (20.21 ± 11.42) μmol/L and (15.11 ± 10.25) μmol/L respectively. The level of serum Hcy in the experimental group was significantly higher than in the control group (t-value of 2.385; p-value of 0.019). The serum Hcy level in the restenosis and the intimal hyperplasia group was (25.72 ± 13.71) μmol/L and (17.35 ± 7.70) μmol/L respectively. The serum Hcy level in the restenosis group was significantly higher than in the intimal hyperplasia group (t-value of 2.215; p-value of 0.033). The level of serum Hcy in the group without a plaque in the stent was (16.30 ± 6.08) μmol/L, whereas in the control group was (15.11 ± 10.25) μmol/L. The no plaque group had a slightly higher serum Hcy level than the control group (t-value of 0.634; p-value of 0.528). All included patients were divided into four quartiles based on the serum Hcy concentration: quartile 1 (8.90–13.20 μmol/L), quartile 2 (13.30–16.45 μmol/L), quartile 3 (16.60–24.25 μmol/L) and quartile 4 (24.30-65.30 μ mol/L). The incidence of ISR was 5, 6.25, 7.5 and 15%, in the 1,2,3 and four quartiles respectively. The serum Hcy level in the experimental group was (20.21 ± 11.42) μmol/L, the severity of in-stent restenosis was (0.25 ± 0.31), (R-value was 0.234; p-value was 0.037), indicating a correlation between serum Hcy and the severity of restenosis (p < 0.05). Taking coronary angiography as the gold standard, a ROC curve analysis was performed on the serum Hcy levels for the experimental group. The area under the curve (AUC) was 0.718 (95% CI 0.585-0.854, p < 0.001), indicating that the serum Hcy concentration could predict ISR. On the ROC curve, the best critical value of serum Hcy concentration for predicting ISR was 20.05 μmol/L, with a sensitivity of 45% and specificity of 88.1%. Conclusion: A positive correlation was observed between homocysteine and the severity of restenosis after PCI, The level of Hcy could serve as a predictive biomarker for the severity of ISR.
Collapse
Affiliation(s)
- Jiqiang Guo
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Gao
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mohammad Ahmed
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
| | - Pengfei Dong
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
| | - Yuping Gao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihua Gong
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinwen Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yajie Mao
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhijie Yue
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingli Zheng
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
| | - Jiansheng Li
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianrong Rong
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongnian Zhou
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meiwen An
- College of Biomedical Engineering, Taiyuan University of Technology, Taiyuan, China
- *Correspondence: Meiwen An, ; Linxia Gu, ; Jin Zhang,
| | - Linxia Gu
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, United States
- *Correspondence: Meiwen An, ; Linxia Gu, ; Jin Zhang,
| | - Jin Zhang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- *Correspondence: Meiwen An, ; Linxia Gu, ; Jin Zhang,
| |
Collapse
|
13
|
Clinical Phenotypes of Cardiovascular and Heart Failure Diseases Can Be Reversed? The Holistic Principle of Systems Biology in Multifaceted Heart Diseases. CARDIOGENETICS 2022. [DOI: 10.3390/cardiogenetics12020015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Recent advances in cardiology and biological sciences have improved quality of life in patients with complex cardiovascular diseases (CVDs) or heart failure (HF). Regardless of medical progress, complex cardiac diseases continue to have a prolonged clinical course with high morbidity and mortality. Interventional coronary techniques together with drug therapy improve quality and future prospects of life, but do not reverse the course of the atherosclerotic process that remains relentlessly progressive. The probability of CVDs and HF phenotypes to reverse can be supported by the advances made on the medical holistic principle of systems biology (SB) and on artificial intelligence (AI). Studies on clinical phenotypes reversal should be based on the research performed in large populations of patients following gathering and analyzing large amounts of relative data that embrace the concept of complexity. To decipher the complexity conundrum, a multiomics approach is needed with network analysis of the biological data. Only by understanding the complexity of chronic heart diseases and explaining the interrelationship between different interconnected biological networks can the probability for clinical phenotypes reversal be increased.
Collapse
|
14
|
Cross B, Turner R, Pirmohamed M. Polygenic risk scores: An overview from bench to bedside for personalised medicine. Front Genet 2022; 13:1000667. [PMID: 36437929 PMCID: PMC9692112 DOI: 10.3389/fgene.2022.1000667] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
Since the first polygenic risk score (PRS) in 2007, research in this area has progressed significantly. The increasing number of SNPs that have been identified by large scale GWAS analyses has fuelled the development of a myriad of PRSs for a wide variety of diseases and, more recently, to PRSs that potentially identify differential response to specific drugs. PRSs constitute a composite genomic biomarker and potential applications for PRSs in clinical practice encompass risk prediction and disease screening, early diagnosis, prognostication, and drug stratification to improve efficacy or reduce adverse drug reactions. Nevertheless, to our knowledge, no PRSs have yet been adopted into routine clinical practice. Beyond the technical considerations of PRS development, the major challenges that face PRSs include demonstrating clinical utility and circumnavigating the implementation of novel genomic technologies at scale into stretched healthcare systems. In this review, we discuss progress in developing disease susceptibility PRSs across multiple medical specialties, development of pharmacogenomic PRSs, and future directions for the field.
Collapse
Affiliation(s)
- Benjamin Cross
- The Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Richard Turner
- The Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Munir Pirmohamed
- The Wolfson Centre for Personalised Medicine, Institute of Systems, Molecular and Integrative Biology, Faculty of Health & Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
15
|
Shakarami A. Incidence of Restenosis Following Rapamycin or Paclitaxeleluting Stent in Coronary Stent Implantation. Cardiovasc Hematol Disord Drug Targets 2021; 21:196-201. [PMID: 34886782 DOI: 10.2174/1871529x21666211209115126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/02/2021] [Accepted: 11/29/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND & OBJECTIVES Coronary artery disease (CAD) is chiefly characterized by atherosclerosis and plaque formation in coronary arteries. The aim of this study was to evaluate the correlation of coronary anatomy as a predictor of restenosis and stent thrombosis in coronary artery disease (CAD) patients 5 years after percutaneous coronary intervention (PCI). METHODS In this prospective study, 1070 patients with stent restenosis or stent thrombosis over past 5 years were enrolled. Coronary angiography was performed to evaluate coronary restenosis and stent thrombosis 5 years after PCI. Stent restenosis was defined as >50% angiographic in-stent lumen reduction. Stent thrombosis was defined as sudden complete occlusion of stent presenting with acute myocardial infarction in that territory. Demographic data, clinical features and anatomic factors were prospectively reviewed. Baseline, procedural, and post-procedural characteristics of patients were recorded for analysis. RESULTS Among demographic characteristics, cardiovascular risk factors (hypertension and diabetes mellitus) and anatomic factors were predictive risk factors for restenosis/thrombosis, p=0.001. The most common site for stent restenosis was proximal to the mid part of the LAD artery, followed by RCA and LCX. A greater diameter of LCX, a greater angle of LM-LAD than LM-LCX and left dominancy increase the incidence of LAD stent restenosis/thrombosis. In this study, the least common restenosis/thrombosis rate in relation to the total number of PCI was in the Ramus intermedius artery. CONCLUSION The outcomes of the study indicated that anatomic factors can predict increased risk of restenosis among CAD patients who underwent PCI.
Collapse
Affiliation(s)
- Amir Shakarami
- Department of Cardiology, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad. Iran
| |
Collapse
|
16
|
Makkos A, Ágg B, Petrovich B, Varga ZV, Görbe A, Ferdinandy P. Systematic review and network analysis of microRNAs involved in cardioprotection against myocardial ischemia/reperfusion injury and infarction: Involvement of redox signalling. Free Radic Biol Med 2021; 172:237-251. [PMID: 33965565 DOI: 10.1016/j.freeradbiomed.2021.04.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/31/2021] [Accepted: 04/27/2021] [Indexed: 01/12/2023]
Abstract
Although myocardial ischemia-reperfusion injury (I/R) and its pathological consequences are the leading cause of morbidity and mortality worldwide, cardioprotective therapeutics are still not on the market. Oxidative stress, a major contributing factor to myocardial I/R, changes transcription of coding and non-coding RNAs, alters post-transcriptional modulations, and regulate protein function. MicroRNA (miRNA) expression can be altered by oxidative stress and microRNAs may also regulate cytoprotective mechanisms and exert cardioprotection againts I/R. Transcriptomic analysis of I/R and oxidative stress-induced alterations followed by microRNA-mRNA target interaction network analysis may reveal microRNAs and their mRNA targets that may play a role in cardioprotection and serve as microRNA therapeutics or novel molecular targets for further drug development. Here we provide a summary of a systematic literature review and in silico molecular network analysis to reveal important cardioprotective microRNAs and their molecular targets that may provide cardioprotection via regulation of redox signalling.
Collapse
Affiliation(s)
- András Makkos
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary.
| | - Bence Ágg
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary; Pharmahungary Group, 6722, Szeged, Hungary.
| | - Balázs Petrovich
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary.
| | - Zoltán V Varga
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; HCEMM-SU Cardiometabolic Immunology Research Group, 1089, Budapest, Hungary.
| | - Anikó Görbe
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary; Pharmahungary Group, 6722, Szeged, Hungary.
| | - Péter Ferdinandy
- Semmelweis University, Department of Pharmacology and Pharmacotherapy, 1089, Budapest, Hungary; MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1089, Budapest, Hungary; Pharmahungary Group, 6722, Szeged, Hungary.
| |
Collapse
|
17
|
Liang T, Liang A, Zhang X, Wang Q, Wu H, He J, Jin T. The association study between CYP20A1, CYP4F2, CYP2D6 gene polymorphisms and coronary heart disease risk in the Han population in southern China. Genes Genomics 2021; 44:1125-1135. [PMID: 34302632 DOI: 10.1007/s13258-021-01125-9] [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: 11/11/2020] [Accepted: 06/16/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Coronary heart disease (CHD) is a disease that seriously harms human health. Genetic factors seriously affect the CHD susceptibility. The CYP20A1, CYP4F2 and CYP2D6 are important drug metabolism enzymes in the human body. OBJECTIVE We aimed to explore the association between CYP20A1, CYP4F2, CYP2D6 single nucleotide polymorphisms (SNPs) and CHD risk in the Chinese Southern Han population. METHODS Based on the 'case-control' experimental design (505 cases and 508 controls), we conducted an association study between 5 candidate SNPs selected from CYP20A1 (rs2043449), CYP4F2 (rs2108622, rs3093106, rs309310), CYP2D6 (rs1065852) and CHD risk. Logistic regression was used to analyze the CHD susceptibility under different genetic models. Multi-factor dimensionality reduction (MDR) was used to analyze the interaction of 'SNP-SNP' in CHD risk. RESULTS Our results showed that under multiple genetic models, CYP2D6 rs1065852 significantly increased the CHD risk in these participants who are ≤ 60 years old (OR 1.40, CI 1.07-1.82, p = 0.013), smokers (OR 1.40, CI 1.02-1.93, p = 0.039), or have family history (OR 1.24, CI 1.02-1.51, p = 0.035). CYP4F2 SNPs rs2108622 (OR 0.63, CI 0.43-0.93, p = 0.020), rs3093106 (OR 0.52, CI 0.29-0.92, p = 0.023), and rs309310 (OR 0.55, CI 0.31-0.96, p = 0.033) were potentially associated with the course of CHD patients. CONCLUSION Our study found that CY2D6 rs1065852 has an outstanding and significant association with increased CHD risk. Our study provided data supplements for CHD genetic susceptibility loci, and also provided a new and valuable reference for CHD drug treatment.
Collapse
Affiliation(s)
- Tiebiao Liang
- Department of Cardiovascular, People's Hospital of Wanning, The First Affiliated Hospital of Chongqing Medical University, Wanning, 571500, Hainan, China
| | - Anshan Liang
- Department of Cardiovascular, People's Hospital of Wanning, The First Affiliated Hospital of Chongqing Medical University, Wanning, 571500, Hainan, China
| | - Xianbo Zhang
- Department of Cardiovascular, People's Hospital of Wanning, The First Affiliated Hospital of Chongqing Medical University, Wanning, 571500, Hainan, China
| | - Qi Wang
- Department of General Practice, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou, 570208, Hainan, China
| | - Haiqing Wu
- Department of Cardiovascular, People's Hospital of Wanning, The First Affiliated Hospital of Chongqing Medical University, Wanning, 571500, Hainan, China
| | - Jun He
- Department of Cardiovascular, People's Hospital of Wanning, The First Affiliated Hospital of Chongqing Medical University, Wanning, 571500, Hainan, China
| | - Tianbo Jin
- Key Laboratory of Molecular Mechanism and Intervention Research for Plateau Diseases of Tibet Autonomous Region, School of Medicine, Xizang Minzu University, Xianyang, 712082, Shaanxi, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China (Northwest University), Ministry of Education, School of Life Sciences, Northwest University, Xi'an, 710069, Shaanxi, China.
| |
Collapse
|
18
|
Roberts R, Fair J. A Less than Provocative Approach for the Primary Prevention of CAD. J Cardiovasc Transl Res 2021; 15:95-102. [PMID: 34128181 DOI: 10.1007/s12265-021-10144-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/31/2021] [Indexed: 12/13/2022]
Abstract
Coronary artery disease (CAD) risk increases in proportion to the magnitude and duration of exposure to plasma low-density lipoprotein cholesterol (LDL-C), doubling every additional decade of exposure. Early primary prevention is three times more effective than initiated later. Several clinical trials show plasma LDL-C of 15-40 mg/dL is more effective and equally safe as the Current Cardiovascular Clinical Practice Guidelines (CCCPG) recommended target of 70mg/dL. The cholesterol in the blood is the excess synthesized by the cells and secreted into the blood for disposal in the liver. The CCCPG is inadequate since traditional risk factors (TRF) are not detectable until the sixth and seventh decade. The genetic risk score (GRS) evaluated in 1 million individuals as a risk stratifier for CAD is superior to TRF. Genetic risk for CAD was reduced by 30-50% by statin therapy, PCSK9 inhibitors, and lifestyle changes. The GRS does not change during one's lifetime and is inexpensive. Incorporating genetic risk stratification into CCCPG would induce a paradigm shift in the primary prevention of CAD.
Collapse
Affiliation(s)
- Robert Roberts
- College of Medicine, The University of Arizona, Phoenix, USA. .,School of Medicine, Creighton University, Phoenix, USA. .,Dignity Health, St. Joseph's Hospital and Medical Center, 500 W Thomas Rd, Phoenix, AZ, 85013, USA.
| | - Jacques Fair
- College of Medicine, The University of Arizona, Phoenix, USA.,School of Medicine, Creighton University, Phoenix, USA.,Dignity Health, St. Joseph's Hospital and Medical Center, 500 W Thomas Rd, Phoenix, AZ, 85013, USA
| |
Collapse
|
19
|
Mendonça MI, Henriques E, Borges S, Sousa AC, Pereira A, Santos M, Temtem M, Freitas S, Monteiro J, Sousa JA, Rodrigues R, Guerra G, dos Reis RP. Genetic information improves the prediction of major adverse cardiovascular events in the GENEMACOR population. Genet Mol Biol 2021; 44:e20200448. [PMID: 34137427 PMCID: PMC8201463 DOI: 10.1590/1678-4685-gmb-2020-0448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/04/2021] [Indexed: 11/21/2022] Open
Abstract
The inclusion of a genetic risk score (GRS) can modify the risk prediction of coronary artery disease (CAD), providing an advantage over the use of traditional models. The predictive value of the genetic information on the recurrence of major adverse cardiovascular events (MACE) remains controversial. A total of 33 genetic variants previously associated with CAD were genotyped in 1587 CAD patients from the GENEMACOR study. Of these, 18 variants presented an hazard ratio >1, so they were selected to construct a weighted GRS (wGRS). MACE discrimination and reclassification were evaluated by C-Statistic, Net Reclassification Index and Integrated Discrimination Improvement methodologies. After the addition of wGRS to traditional predictors, the C-index increased from 0.566 to 0.572 (p=0.0003). Subsequently, adding wGRS to traditional plus clinical risk factors, this model slightly improved from 0.620 to 0.622 but with statistical significance (p=0.004). NRI showed that 17.9% of the cohort was better reclassified when the primary model was associated with wGRS. The Kaplan-Meier estimator showed that, at 15-year follow-up, the group with a higher number of risk alleles had a significantly higher MACE occurrence (p=0.011). In CAD patients, wGRS improved MACE risk prediction, discrimination and reclassification over the conventional factors, providing better cost-effective therapeutic strategies.
Collapse
Affiliation(s)
- Maria Isabel Mendonça
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Eva Henriques
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Sofia Borges
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Ana Célia Sousa
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Andreia Pereira
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Marina Santos
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Margarida Temtem
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Sónia Freitas
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Joel Monteiro
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - João Adriano Sousa
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Ricardo Rodrigues
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | - Graça Guerra
- Hospital Central do Funchal, Unidade de Investigação, Serviço de
Saúde da Região, SESARAM, EPERAM, Funchal, Portugal
| | | |
Collapse
|
20
|
Roberts R, Chang CC, Hadley T. Genetic Risk Stratification: A Paradigm Shift in Prevention of Coronary Artery Disease. ACTA ACUST UNITED AC 2021; 6:287-304. [PMID: 33778213 PMCID: PMC7987546 DOI: 10.1016/j.jacbts.2020.09.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/08/2020] [Accepted: 09/13/2020] [Indexed: 12/12/2022]
Abstract
CAD is a pandemic that can be prevented. Conventional risk factors are inadequate to detect who is at risk early in the asymptomatic stage. Genetic risk for CAD can be determined at birth, and those at highest genetic risk have been shown to respond to lifestyle changes and statin therapy with a 40% to 50% reduction in cardiac events. Genetic risk stratification for CAD should be brought to the bedside in an attempt to prevent this pandemic disease.
Coronary artery disease (CAD) is a pandemic disease that is highly preventable as shown by secondary prevention. Primary prevention is preferred knowing that 50% of the population can expect a cardiac event in their lifetime. Risk stratification for primary prevention using the American Heart Association/American College of Cardiology predicted 10-year risk based on conventional risk factors for CAD is less than optimal. Conventional risk factors such as hypertension, cholesterol, and age are age-dependent and not present until the sixth or seventh decade of life. The genetic risk score (GRS), which is estimated from the recently discovered genetic variants predisposed to CAD, offers a potential solution to this dilemma. The GRS, which is derived from genotyping the population with a microarray containing these genetic risk variants, has indicated that genetic risk stratification based on the GRS is superior to that of conventional risk factors in detecting those at high risk and who would benefit most from statin therapy. Studies performed in >1 million individuals confirmed genetic risk stratification is superior and primarily independent of conventional risk factors. Prospective clinical trials based on risk stratification for CAD using the GRS have shown lifestyle changes, physical activity, and statin therapy are associated with 40% to 50% reduction in cardiac events in the high genetic risk group (20%). Genetic risk stratification has the advantage of being innate to an individual’s DNA, and because DNA does not change in a lifetime, it is independent of age. Genetic risk stratification is inexpensive and can be performed worldwide, providing risk analysis at any age and thus has the potential to revolutionize primary prevention.
Collapse
Key Words
- ACC, American College of Cardiology
- AHA, American Heart Association
- ANRIL, antisense non-coding RNA in the INK4 Locust
- CAD, coronary artery disease
- GRS, genetic risk score
- GWAS, genome-wide association study
- LDL-C, low-density lipoprotein cholesterol
- MR, Mendelian randomization
- SNP, single nucleotide polymorphism
- bp, base pair
- cardiovascular genetics
- coronary artery disease
- genetic risk score for CAD
- genome-wide association studies
- prevention of CAD
Collapse
Affiliation(s)
- Robert Roberts
- Department of Medicine, Dignity Health at St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | - Chih Chao Chang
- Department of Medicine, Dignity Health at St. Joseph's Hospital and Medical Center, Phoenix, Arizona, USA
| | | |
Collapse
|
21
|
Zhang J, Zhang Y, He X, Wang S, Pang S, Yan B. TFEB Gene Promoter Variants Effect on Gene Expression in Acute Myocardial Infarction. Front Cell Dev Biol 2021; 9:630279. [PMID: 33732699 PMCID: PMC7959723 DOI: 10.3389/fcell.2021.630279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/26/2021] [Indexed: 11/23/2022] Open
Abstract
Autophagy is involved in many physiological processes. Transcription factor EB (TFEB) is a master regulator of autophagy and coordinates the expression of autophagic proteins, lysosomal hydrolases, and lysosomal membrane proteins. Though autophagy has been implicated in several human diseases, little is known regarding TFEB gene expression and regulation in the process. Since dysfunctional autophagy plays critical roles in acute myocardial infarction (AMI), dysregulated TFEB gene expression may be associated with AMI by regulating autophagy. In this study, the TFEB gene promoter was genetically and functionally analyzed in AMI patients (n = 352) and ethnic-matched controls (n = 337). A total of fifteen regulatory variants of the TFEB gene, including eight single-nucleotide polymorphisms (SNPs), were identified in this population. Among these, six regulatory variants [g.41737274T>C (rs533895008), g.41737144A>G, g.41736987C > T (rs760293138), g.41736806C > T (rs748537297), g.41736635T > C (rs975050638), and g.41736544C > T] were only identified in AMI patients. These regulatory variants significantly altered the transcriptional activity of the TFEB gene promoter. Further electrophoretic mobility shift assay revealed that three of the variants evidently affected the binding of transcription factors. Therefore, this study identified novel TFEB gene regulatory variants which affect the gene expression. These TFEB gene regulatory variants may contribute to AMI development as a rare risk factor.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Medicine, Shandong University School of Medicine, Jinan, China
| | - Yexin Zhang
- Department of Medicine, Shandong University School of Medicine, Jinan, China
| | - Xiaohui He
- Department of Medicine, Shandong University School of Medicine, Jinan, China
| | - Shuai Wang
- Department of Medicine, Shandong University School of Medicine, Jinan, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,The Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,The Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| |
Collapse
|
22
|
Wang X, Meng H, Ruan J, Chen W, Meng F. Low G0S2 gene expression levels in peripheral blood may be a genetic marker of acute myocardial infarction in patients with stable coronary atherosclerotic disease: A retrospective clinical study. Medicine (Baltimore) 2021; 100:e23468. [PMID: 33545927 PMCID: PMC7837852 DOI: 10.1097/md.0000000000023468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 11/02/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The G0/G1 switch 2 (G0S2) gene is closely related to lipolysis, cell proliferation, apoptosis, oxidative phosphorylation, and the development of a variety of tumors. The aim of the present study was to expand the sample size to confirm the relationship between the expression of the G0S2 gene in peripheral blood and acute myocardial infarction (AMI) based on previous gene chip results. METHODS Three hundred patients were initially selected, of which 133 were excluded in accordance with the exclusion criteria. Peripheral blood leukocytes were collected from 92 patients with AMI and 75 patients with stable coronary atherosclerotic disease (CAD). mRNA expression levels of G0S2 in peripheral blood leukocytes was measured by RT-PCR, and protein expression levels by Western blot analysis. The results of these assays in the 2 groups were compared. RESULTS mRNA expression levels of GOS2 in the peripheral blood leukocytes of patients with AMI were 0.41-fold lower than those of patients with stable CAD (P < .05), and GOS2 protein expression levels were 0.45-fold lower. Multivariate logistic regression analysis indicated that low expression levels of the G0S2 gene increased the risk of AMI by 2.08-fold in stable CAD patients. CONCLUSIONS G0S2 gene expression in the peripheral blood leukocytes of AMI patients was lower than that of stable CAD patients. Low G0S2 gene expression in peripheral blood leukocytes is an independent risk factor for AMI in stable CAD patients.
Collapse
|
23
|
Roberts R. Cholesterol Surprisingly Also Induces Ventricular Hypertrophy. J Am Coll Cardiol 2020; 76:2489-2491. [PMID: 33213728 DOI: 10.1016/j.jacc.2020.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 12/23/2022]
Affiliation(s)
- Robert Roberts
- University of Arizona, College of Medicine, Phoenix, Arizona; St. Joseph's Hospital and Medical Center, Dignity Health, Phoenix, Arizona.
| |
Collapse
|
24
|
Whetton AD, Preston GW, Abubeker S, Geifman N. Proteomics and Informatics for Understanding Phases and Identifying Biomarkers in COVID-19 Disease. J Proteome Res 2020; 19:4219-4232. [PMID: 32657586 PMCID: PMC7384384 DOI: 10.1021/acs.jproteome.0c00326] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Indexed: 02/07/2023]
Abstract
The emergence of novel coronavirus disease 2019 (COVID-19), caused by the SARS-CoV-2 coronavirus, has necessitated the urgent development of new diagnostic and therapeutic strategies. Rapid research and development, on an international scale, has already generated assays for detecting SARS-CoV-2 RNA and host immunoglobulins. However, the complexities of COVID-19 are such that fuller definitions of patient status, trajectory, sequelae, and responses to therapy are now required. There is accumulating evidence-from studies of both COVID-19 and the related disease SARS-that protein biomarkers could help to provide this definition. Proteins associated with blood coagulation (D-dimer), cell damage (lactate dehydrogenase), and the inflammatory response (e.g., C-reactive protein) have already been identified as possible predictors of COVID-19 severity or mortality. Proteomics technologies, with their ability to detect many proteins per analysis, have begun to extend these early findings. To be effective, proteomics strategies must include not only methods for comprehensive data acquisition (e.g., using mass spectrometry) but also informatics approaches via which to derive actionable information from large data sets. Here we review applications of proteomics to COVID-19 and SARS and outline how pipelines involving technologies such as artificial intelligence could be of value for research on these diseases.
Collapse
Affiliation(s)
- Anthony D. Whetton
- Stoller
Biomarker Discovery Centre, Faculty of Biology Medicine and Health
(FBMH), University of Manchester, Manchester M20 4GJ, United Kingdom
- Stem
Cell and Leukaemia Proteomics Laboratory, Manchester Cancer Research
Centre, University of Manchester, Manchester M13 9PL, United Kingdom
- Manchester
National Institute for Health Biomedical Research Centre, Manchester M13 9WL, United Kingdom
| | - George W. Preston
- Stoller
Biomarker Discovery Centre, Faculty of Biology Medicine and Health
(FBMH), University of Manchester, Manchester M20 4GJ, United Kingdom
- Stem
Cell and Leukaemia Proteomics Laboratory, Manchester Cancer Research
Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Semira Abubeker
- Stoller
Biomarker Discovery Centre, Faculty of Biology Medicine and Health
(FBMH), University of Manchester, Manchester M20 4GJ, United Kingdom
- Stem
Cell and Leukaemia Proteomics Laboratory, Manchester Cancer Research
Centre, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Nophar Geifman
- Centre
for Health Informatics, FBMH, University
of Manchester, Manchester M13 9PL, United Kingdom
| |
Collapse
|
25
|
Wang H, Liu Z, Shao J, Jiang M, Lu X, Lin L, Wang L, Xu Q, Zhang H, Li X, Zhou J, Chen Y, Zhang R. Pathogenesis of premature coronary artery disease: Focus on risk factors and genetic variants. Genes Dis 2020; 9:370-380. [PMID: 35224153 PMCID: PMC8843894 DOI: 10.1016/j.gendis.2020.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/17/2020] [Accepted: 11/04/2020] [Indexed: 11/24/2022] Open
Abstract
The development of premature coronary artery disease (PCAD) is dependent on both genetic predisposition and traditional risk factors. Strategies for unraveling the genetic basis of PCAD have evolved with the advent of modern technologies. Genome-wide association studies (GWASs) have identified a considerable number of common genetic variants that are associated with PCAD. Most of these genetic variants are attributable to lipid and blood pressure-related single-nucleotide polymorphisms (SNPs). The genetic variants that predispose individuals to developing PCAD may depend on race and ethnicity. Some characteristic genetic variants have been identified in Chinese populations. Although translating this genetic knowledge into clinical applications is still challenging, these genetic variants can be used for CAD phenotype identification, genetic prediction and therapy. In this article we will provide a comprehensive review of genetic variants detected by GWASs that are predicted to contribute to the development of PCAD. We will highlight recent findings regarding CAD-related genetic variants in Chinese populations and discuss the potential clinical utility of genetic variants for preventing and managing PCAD.
Collapse
|
26
|
Alizadehsani R, Khosravi A, Roshanzamir M, Abdar M, Sarrafzadegan N, Shafie D, Khozeimeh F, Shoeibi A, Nahavandi S, Panahiazar M, Bishara A, Beygui RE, Puri R, Kapadia S, Tan RS, Acharya UR. Coronary artery disease detection using artificial intelligence techniques: A survey of trends, geographical differences and diagnostic features 1991-2020. Comput Biol Med 2020; 128:104095. [PMID: 33217660 DOI: 10.1016/j.compbiomed.2020.104095] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 10/24/2020] [Accepted: 10/24/2020] [Indexed: 02/06/2023]
Abstract
While coronary angiography is the gold standard diagnostic tool for coronary artery disease (CAD), but it is associated with procedural risk, it is an invasive technique requiring arterial puncture, and it subjects the patient to radiation and iodinated contrast exposure. Artificial intelligence (AI) can provide a pretest probability of disease that can be used to triage patients for angiography. This review comprehensively investigates published papers in the domain of CAD detection using different AI techniques from 1991 to 2020, in order to discern broad trends and geographical differences. Moreover, key decision factors affecting CAD diagnosis are identified for different parts of the world by aggregating the results from different studies. In this study, all datasets that have been used for the studies for CAD detection, their properties, and achieved performances using various AI techniques, are presented, compared, and analyzed. In particular, the effectiveness of machine learning (ML) and deep learning (DL) techniques to diagnose and predict CAD are reviewed. From PubMed, Scopus, Ovid MEDLINE, and Google Scholar search, 500 papers were selected to be investigated. Among these selected papers, 256 papers met our criteria and hence were included in this study. Our findings demonstrate that AI-based techniques have been increasingly applied for the detection of CAD since 2008. AI-based techniques that utilized electrocardiography (ECG), demographic characteristics, symptoms, physical examination findings, and heart rate signals, reported high accuracy for the detection of CAD. In these papers, the authors ranked the features based on their assessed clinical importance with ML techniques. The results demonstrate that the attribution of the relative importance of ML features for CAD diagnosis is different among countries. More recently, DL methods have yielded high CAD detection performance using ECG signals, which drives its burgeoning adoption.
Collapse
Affiliation(s)
- Roohallah Alizadehsani
- Institute for Intelligent Systems Research and Innovations (IISRI), Deakin University, Geelong, Australia
| | - Abbas Khosravi
- Institute for Intelligent Systems Research and Innovations (IISRI), Deakin University, Geelong, Australia
| | - Mohamad Roshanzamir
- Department of Engineering, Fasa Branch, Islamic Azad University, Post Box No 364, Fasa, Fars, 7461789818, Iran
| | - Moloud Abdar
- Institute for Intelligent Systems Research and Innovations (IISRI), Deakin University, Geelong, Australia
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Khorram Ave, Isfahan, Iran; Faculty of Medicine, SPPH, University of British Columbia, Vancouver, BC, Canada.
| | - Davood Shafie
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fahime Khozeimeh
- Institute for Intelligent Systems Research and Innovations (IISRI), Deakin University, Geelong, Australia
| | - Afshin Shoeibi
- Computer Engineering Department, Ferdowsi University of Mashhad, Mashhad, Iran; Faculty of Electrical and Computer Engineering, Biomedical Data Acquisition Lab, K. N. Toosi University of Technology, Tehran, Iran
| | - Saeid Nahavandi
- Institute for Intelligent Systems Research and Innovations (IISRI), Deakin University, Geelong, Australia
| | - Maryam Panahiazar
- Institute for Computational Health Sciences, University of California, San Francisco, USA
| | - Andrew Bishara
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, USA
| | - Ramin E Beygui
- Cardiovascular Surgery Division, Department of Surgery, University of California, San Francisco, CA, USA
| | - Rishi Puri
- Department of Cardiovascular Medicine, Cleveland Clinic, OH, USA
| | - Samir Kapadia
- Department of Cardiovascular Medicine, Cleveland Clinic, OH, USA
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore
| | - U Rajendra Acharya
- Department of Electronics and Computer Engineering, Ngee Ann Polytechnic, Singapore; Department of Biomedical Engineering, School of Science and Technology, Singapore University of Social Sciences, Singapore; Department of Bioinformatics and Medical Engineering, Asia University, Taiwan
| |
Collapse
|
27
|
Gooding HC, Gidding SS, Moran AE, Redmond N, Allen NB, Bacha F, Burns TL, Catov JM, Grandner MA, Harris KM, Johnson HM, Kiernan M, Lewis TT, Matthews KA, Monaghan M, Robinson JG, Tate D, Bibbins-Domingo K, Spring B. Challenges and Opportunities for the Prevention and Treatment of Cardiovascular Disease Among Young Adults: Report From a National Heart, Lung, and Blood Institute Working Group. J Am Heart Assoc 2020; 9:e016115. [PMID: 32993438 PMCID: PMC7792379 DOI: 10.1161/jaha.120.016115] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Improvements in cardiovascular disease (CVD) rates among young adults in the past 2 decades have been offset by increasing racial/ethnic and gender disparities, persistence of unhealthy lifestyle habits, overweight and obesity, and other CVD risk factors. To enhance the promotion of cardiovascular health among young adults 18 to 39 years old, the medical and broader public health community must understand the biological, interpersonal, and behavioral features of this life stage. Therefore, the National Heart, Lung, and Blood Institute, with support from the Office of Behavioral and Social Science Research, convened a 2-day workshop in Bethesda, Maryland, in September 2017 to identify research challenges and opportunities related to the cardiovascular health of young adults. The current generation of young adults live in an environment undergoing substantial economic, social, and technological transformations, differentiating them from prior research cohorts of young adults. Although the accumulation of clinical and behavioral risk factors for CVD begins early in life, and research suggests early risk is an important determinant of future events, few trials have studied prevention and treatment of CVD in participants <40 years old. Building an evidence base for CVD prevention in this population will require the engagement of young adults, who are often disconnected from the healthcare system and may not prioritize long-term health. These changes demand a repositioning of existing evidence-based treatments to accommodate new sociotechnical contexts. In this article, the authors review the recent literature and current research opportunities to advance the cardiovascular health of today's young adults.
Collapse
Affiliation(s)
- Holly C Gooding
- Division of General Pediatrics and Adolescent Medicine Emory UniversityChildren's Healthcare of Atlanta Atlanta GA
| | | | - Andrew E Moran
- Division of General Medicine Columbia University New York NY
| | | | - Norrina B Allen
- Department of Preventive Medicine Northwestern University Feinberg School of Medicine Chicago IL
| | - Fida Bacha
- Division of Pediatric Endocrinology and Diabetes Texas Children's HospitalBaylor College of Medicine Houston TX
| | - Trudy L Burns
- Department of Epidemiology University of Iowa Iowa City IA
| | - Janet M Catov
- Department of Obstetrics, Gynecology & Reproductive Sciences Department of Epidemiology University of Pittsburgh Pittsburgh PA
| | | | | | - Heather M Johnson
- Blechman Center for Specialty Care and Preventive Cardiology Boca Raton Regional Hospital/Baptist Health South Florida Boca Raton FL
| | - Michaela Kiernan
- Department of Medicine Stanford University School of Medicine Stanford CA
| | - Tené T Lewis
- Department of Epidemiology Emory University, Children's Healthcare of Atlanta Atlanta GA
| | | | - Maureen Monaghan
- Department of Psychiatry and Behavioral Sciences Department of Pediatrics Children's National Health System George Washington University School of Medicine Washington DC
| | | | - Deborah Tate
- Department of Sociology University of North Carolina at Chapel Hill Chapel Hill NC
| | - Kirsten Bibbins-Domingo
- Department of Epidemiology and Biostatistics University of California San Francisco San Francisco CA
| | - Bonnie Spring
- Department of Preventive Medicine Northwestern University Feinberg School of Medicine Chicago IL
| |
Collapse
|
28
|
Roberts R, Chang CC. A Journey through Genetic Architecture and Predisposition of Coronary Artery Disease. Curr Genomics 2020; 21:382-398. [PMID: 33093801 PMCID: PMC7536803 DOI: 10.2174/1389202921999200630145241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/18/2020] [Accepted: 05/26/2020] [Indexed: 01/14/2023] Open
Abstract
Introduction To halt the spread of coronary artery disease (CAD), the number one killer in the world, requires primary prevention. Fifty percent of all Americans are expected to experience a cardiac event; the challenge is identifying those at risk. 40 to 60% of predisposition to CAD is genetic. The first genetic risk variant, 9p21, was discovered in 2007. Genome-Wide Association Studies has since discovered hundreds of genetic risk variants. The genetic burden for CAD can be expressed as a single number, Genetic Risk Score (GRS). Assessment of GRS to risk stratify for CAD was superior to conventional risk factors in several large clinical trials assessing statin therapy, and more recently in a population of nearly 500,000 (UK Biobank). Studies were performed based on prospective genetic risk stratification for CAD. These studies showed that a favorable lifestyle was associated with a 46% reduction in cardiac events and programmed exercise, a 50% reduction in cardiac events. Genetic risk score is superior to conventional risk factors, and is markedly attenuated by lifestyle changes and drug therapy. Genetic risk can be determined at birth or any time thereafter. Conclusion Utilizing the GRS to risk stratify young, asymptomatic individuals could provide a paradigm shift in the primary prevention of CAD and significantly halt its spread.
Collapse
Affiliation(s)
- Robert Roberts
- 1Cardiovascular Genomics & Genetics, University of Arizona, College of Medicine, Phoenix, AZ, USA; 2Cardiovascular Genomics & Genetics, Phoenix, AZ, USA
| | - Chih Chao Chang
- 1Cardiovascular Genomics & Genetics, University of Arizona, College of Medicine, Phoenix, AZ, USA; 2Cardiovascular Genomics & Genetics, Phoenix, AZ, USA
| |
Collapse
|
29
|
Rincón LM, Sanmartín M, Alonso GL, Rodríguez JA, Muriel A, Casas E, Navarro M, Carbonell A, Lázaro C, Fernández S, González P, Rodríguez M, Jiménez-Mena M, Fernández-Golfín C, Esteban A, García-Bermejo ML, Zamorano JL. Una puntuación de riesgo genético predice recurrencias en pacientes jóvenes con infarto agudo de miocardio. Rev Esp Cardiol 2020. [DOI: 10.1016/j.recesp.2019.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
30
|
Li F, Hu S, Zhou X, Mei X, Zhou Y. Association Between R353Q (rs6046) Polymorphism in Factor VII with Coronary Heart Disease. Int Heart J 2020; 61:641-650. [PMID: 32684587 DOI: 10.1536/ihj.19-219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A number of studies have showed the relationship between R353Q (rs6046) polymorphism in factor VII gene and coronary heart disease (CHD). However, the results remain controversial due to the limitations of the research objects and small sample size of individual study. We conducted this meta-analysis to validate the association between R353Q (rs6046) polymorphism and the risk of CHD.The relevant data was collected up to March 25, 2019 from PubMed, Web of Science, CNKI, and Wanfang databases. We examined all eligible studies using the Newcastle-Ottawa Quality Assessment Scale (NOS). The odds ratio (OR) and its corresponding 95% confidence interval (CI) were adopted to evaluate the relationship between the R353Q (rs6046) polymorphism and CHD. Stata version 14.0 (Stata Corporation, USA) was used in all statistical tests.There were at least 28 eligible studies, including 14626 cases and 17994 controls, included in our meta-analysis. R353Q (rs6046) polymorphism was associated with the reduced risk of CHD in four genetic models: allele model (Q versus R: OR = 0.79, 95% CI: 0.69 to 0.90, P < 0.001, I2 = 56.4%), homozygote (co-dominant) model (QQ versus RR: OR = 0.72, 95% CI = 0.58 to 0.92, P = 0.004, I2 = 5.8%), heterozygote (co-dominant) model (RQ versus RR: OR = 0.71, 95% CI = 0.58 to 0.86, P = 0.001, I2 = 75.4%), and dominant model (RQ+QQ versus RR: OR = 0.74, 95% CI = 0.63 to 0.865, P < 0.001, I2 = 64.1%) excluding recessive model (QQ versus RR+RQ: OR = 0.86, 95% CI = 0.57 to 1.28, P = 0.447, I2 = 51.6%).The results of the current meta-analysis suggested that R353Q (rs6046) polymorphism was associated with the reduced risk of CHD, especially in Asians.
Collapse
Affiliation(s)
- Fei Li
- Department of Cardiology, The First Affiliated Hospital of Soochow University
| | - Shengda Hu
- Department of Cardiology, The First Affiliated Hospital of Soochow University
| | - Xianyong Zhou
- Department of Cardiology, The First Affiliated Hospital of Soochow University
| | - Xiaofei Mei
- Department of Cardiology, The First Affiliated Hospital of Soochow University
| | - Yafeng Zhou
- Department of Cardiology, The First Affiliated Hospital of Soochow University
| |
Collapse
|
31
|
Mendelian randomization in COVID-19: Applications for cardiovascular comorbidities and beyond. EBioMedicine 2020; 57:102847. [PMID: 32574960 PMCID: PMC7305723 DOI: 10.1016/j.ebiom.2020.102847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 01/22/2023] Open
|
32
|
Sharifi-Rad J, Rodrigues CF, Sharopov F, Docea AO, Can Karaca A, Sharifi-Rad M, Kahveci Karıncaoglu D, Gülseren G, Şenol E, Demircan E, Taheri Y, Suleria HAR, Özçelik B, Nur Kasapoğlu K, Gültekin-Özgüven M, Daşkaya-Dikmen C, Cho WC, Martins N, Calina D. Diet, Lifestyle and Cardiovascular Diseases: Linking Pathophysiology to Cardioprotective Effects of Natural Bioactive Compounds. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E2326. [PMID: 32235611 PMCID: PMC7177934 DOI: 10.3390/ijerph17072326] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/12/2022]
Abstract
Heart and blood vessels disorders comprise one of the main causes of death worldwide. Pharmacologically active natural compounds have been used as a complementary therapy in cardiovascular disease around the world in a traditional way. Dietary, natural bioactive compounds, as well as healthy lifestyles, are considered to prevent coronary artery diseases. Pre-clinical and clinical studies reported that consumption of plant-food bioactive derivatives including polyphenolic compounds, peptides, oligosaccharides, vitamins, unsaturated fatty acids possess protective effects on cardiovascular diseases. This review aims to summarize the cardiovascular risk factors, pre-clinical studies and clinical trials related to cardioprotective properties of the plant-food-derived bioactive compounds. Molecular mechanisms by the natural bioactive compounds exert their cardiovascular protective properties have also been highlighted.
Collapse
Affiliation(s)
- Javad Sharifi-Rad
- Zabol Medicinal Plants Research Center, Zabol University of Medical Sciences, Zabol 61615-585, Iran;
| | - Célia F. Rodrigues
- LEPABE—Department of Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, s/n, 4200-465 Porto, Portugal;
| | - Farukh Sharopov
- Department of Pharmaceutical Technology, Avicenna Tajik State Medical University, Rudaki 139, 734003 Dushanbe, Tajikistan;
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Aslı Can Karaca
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak, Istanbul 34469, Turkey; (A.C.K.); (D.K.K.); (B.O.); (K.N.K.); (M.G.-Ö.)
| | - Mehdi Sharifi-Rad
- Department of Medical Parasitology, Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran;
| | - Derya Kahveci Karıncaoglu
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak, Istanbul 34469, Turkey; (A.C.K.); (D.K.K.); (B.O.); (K.N.K.); (M.G.-Ö.)
| | - Gözde Gülseren
- Department of Food Engineering, Chemical and Metallurgical Faculty, Istanbul Technical University, Maslak Istanbul 34469, Turkey; (G.G.); (E.Ş.); (E.D.)
| | - Ezgi Şenol
- Department of Food Engineering, Chemical and Metallurgical Faculty, Istanbul Technical University, Maslak Istanbul 34469, Turkey; (G.G.); (E.Ş.); (E.D.)
| | - Evren Demircan
- Department of Food Engineering, Chemical and Metallurgical Faculty, Istanbul Technical University, Maslak Istanbul 34469, Turkey; (G.G.); (E.Ş.); (E.D.)
| | - Yasaman Taheri
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran 1991953381, Iran;
| | | | - Beraat Özçelik
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak, Istanbul 34469, Turkey; (A.C.K.); (D.K.K.); (B.O.); (K.N.K.); (M.G.-Ö.)
- Bioactive Research & Innovation Food Manufac. Indust. Trade Ltd., Katar Street, Teknokent ARI-3, B110, Sarıyer, Istanbul 34467, Turkey
| | - Kadriye Nur Kasapoğlu
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak, Istanbul 34469, Turkey; (A.C.K.); (D.K.K.); (B.O.); (K.N.K.); (M.G.-Ö.)
| | - Mine Gültekin-Özgüven
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Maslak, Istanbul 34469, Turkey; (A.C.K.); (D.K.K.); (B.O.); (K.N.K.); (M.G.-Ö.)
| | - Ceren Daşkaya-Dikmen
- Pladis TR R&D Department, Kısıklı mah., Ferah cad. Üsküdar İstanbul 34692, Turkey;
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, 30 Gascoigne Road, Hong Kong, China
| | - Natália Martins
- Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| |
Collapse
|
33
|
Vuković Dejanović V, Bogavac-Stanojević N, Kotur Stevuljević J, Vukašinović A, Stefanović A, Kafedzic S, Zdravković M, Ilić I, Hinić S, Cerović M, Stefanović M, Spasojević-Kalimanovska V, Memon L, Nešković AN. Circulating levels of inflammatory parameters pentraxin-3, cyclophilin and heparin-binding epidermal growth factor-like growth factor in patients with ST-elevation myocardial infarction. Scandinavian Journal of Clinical and Laboratory Investigation 2019; 80:66-72. [PMID: 31799884 DOI: 10.1080/00365513.2019.1698058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Inflammatory biomarkers - pentraxin-3 (PTX3), cyclophilin A (CypA) and heparin-binding epidermal growth factor-like growth factor (HB-EGF) were examined in patients with ST-segment elevation myocardial infarction (STEMI) undergoing revascularization with primary percutaneous coronary intervention (pPCI) and stent implanting. Investigated parameters were compared between patients with and without obstructive coronary artery disease (CAD). In addition, their changes were tested in circulation before and immediately after pPCI. The study group consisted of 81 STEMI patients. Patients were classified in the STEMI-CAD group if they had significant obstructive CAD or in MINOCA group if they had no significant stenosis. In STEMI-CAD patients inflammatory parameters were determined prior to and after pPCI intervention. Immediately after pPCI, in STEMI-CAD patients levels of PTX3 were significantly lower (1.52 vs. 2.17 μg/L, p < .001), while the levels of HB-EGF (14.61 vs. 12.03 pg/L, p < .001) and CyPA (15.95 vs. 8.62 μg/L, p < .001) were significantly higher compared to levels before pPCI. STEMI-CAD patients had lower PTX3 values 2.17 μg/L (1.55-5.10 μg/L) than MINOCA patients 5.06 μg/L (2.77-6.7 μg/L), p = .046. Diagnostic accuracy of PTX3 for discrimination MINOCA from STEMI-CAD patients was low (area under receiver operating characteristic curve = 0.770). Evaluation of PTX3 values may be helpful in the understanding of MINOCA aetiology but they couldn't distinguish stenosis severity in STEMI patients. Inflammatory biomarkers significantly changed after pPCI but the possibility of clinical use of these biomarkers needs to be evaluated in a larger prospective study.
Collapse
Affiliation(s)
| | | | - Jelena Kotur Stevuljević
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Vukašinović
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Stefanović
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Srdjan Kafedzic
- Department of Cardiology, Clinical Hospital Center Zemun, Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Marija Zdravković
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Department of Cardiology, Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Ivan Ilić
- Department of Cardiology, Clinical Hospital Center Zemun, Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Saša Hinić
- Department of Cardiology, Clinical Hospital Center Bezanijska kosa, Belgrade, Serbia
| | - Milivoje Cerović
- Department of Cardiology, Clinical Hospital Center Zemun, Belgrade, Serbia
| | - Milica Stefanović
- Department of Cardiology, Clinical Hospital Center Zemun, Belgrade, Serbia
| | | | - Lidija Memon
- Clinical Hospital Center Bezanijska kosa, Clinical Chemistry Laboratory, Belgrade, Serbia
| | - Aleksandar N Nešković
- Department of Cardiology, Clinical Hospital Center Zemun, Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
34
|
Wang S, Zhang J, He X, Zhang Y, Chen J, Su Q, Pang S, Zhang S, Cui Y, Yan B. Identification and functional analysis of genetic variants in TBX5 gene promoter in patients with acute myocardial infarction. BMC Cardiovasc Disord 2019; 19:265. [PMID: 31775637 PMCID: PMC6880377 DOI: 10.1186/s12872-019-1237-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 10/24/2019] [Indexed: 01/09/2023] Open
Abstract
Background Coronary artery disease (CAD), including acute myocardial infarction (AMI), is a common complex disease. Although a great number of genetic loci and variants for CAD have been identified, genetic causes and underlying mechanisms remain largely unclear. Epidemiological studies have revealed that CAD incidence is strikingly higher in patients with congenital heart disease than that in normal population. T-box transcription factors play critical roles in embryonic development. In particular, TBX5 as a dosage-sensitive regulator is required for cardiac development and function. Thus, dysregulated TBX5 gene expression may be involved in CAD development. Methods TBX5 gene promoter was genetically and functionally analysed in large groups of AMI patients (n = 432) and ethnic-matched healthy controls (n = 448). Results Six novel heterozygous DNA sequence variants (DSVs) in the TBX5 gene promoter (g.4100A > G, g.4194G > A, g.4260 T > C, g.4367C > A, g.4581A > G and g.5004G > T) were found in AMI patients, but in none of controls. These DSVs significantly changed the activity of TBX5 gene promoter in cultured cells (P < 0.05). Furthermore, three of the DSVs (g.4100A > G, g.4260 T > C and g.4581A > G) evidently modified the binding sites of unknown transcription factors. Conclusions The DSVs identified in AMI patients may alter TBX5 gene promoter activity and change TBX5 level, contributing to AMI development as a rare risk factor.
Collapse
Affiliation(s)
- Shuai Wang
- Department of Medicine, Shandong University School of Medicine, Jinan, 250012, Shandong, China
| | - Jie Zhang
- Department of Medicine, Shandong University School of Medicine, Jinan, 250012, Shandong, China
| | - Xiaohui He
- Department of Medicine, Shandong University School of Medicine, Jinan, 250012, Shandong, China
| | - Yexin Zhang
- Department of Medicine, Shandong University School of Medicine, Jinan, 250012, Shandong, China
| | - Jing Chen
- Department of Medicine, Shandong University School of Medicine, Jinan, 250012, Shandong, China
| | - Qiang Su
- Department of Medicine, Shandong University School of Medicine, Jinan, 250012, Shandong, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong, China.,Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China
| | - Shufang Zhang
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong, China
| | - Yinghua Cui
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong, China.
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, 272029, Shandong, China. .,Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China. .,Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272029, Shandong, China.
| |
Collapse
|
35
|
Genetic variants of VEGFR-1 gene promoter in acute myocardial infarction. Hum Genomics 2019; 13:56. [PMID: 31744542 PMCID: PMC6862733 DOI: 10.1186/s40246-019-0243-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Coronary artery disease (CAD) including acute myocardial infarction (AMI) is a common complex disease caused by atherosclerosis. Vascular epithelial growth factor receptor-1 (VEGFR-1) stimulates angiogenesis and vascular permeability, and functions as a decoy to sequester VEGF and prevent initiation of intracellular signaling. VEGFR-1 knockout mice exhibit significantly higher mortality due to heart failure, cardiac hypertrophy, and cardiac dysfunction. An evident increase in macrophage infiltration and cardiac fibrosis are also observed after transverse aortic constriction. Therefore, VEGFR-1 gene variants may be involved in CAD. In this study, VEGFR-1 gene promoter was genetically and functionally analyzed in large cohorts of AMI patients and ethnic-matched controls. RESULTS A total of 16 DNA sequence variants (DSVs) including six single-nucleotide polymorphisms (SNPs) were found in the VEGFR-1 gene promoter and 5'-untranslated region. Five novel DSVs and one SNP were only identified in AMI patients group. These DSVs and SNP significantly altered the transcriptional activity of the VEGFR-1 gene promoter in both HEK-293 and H9c2 cells (P < 0.05). Further electrophoretic mobility shift assay indicated that the DSVs and SNPs evidently affected the binding of transcription factors. CONCLUSIONS The genetic variants in VEGFR-1 gene identified in AMI patients may alter the transcriptional activity of the VEGFR-1 gene promoter and change VEGFR-1 level, contributing to AMI development.
Collapse
|
36
|
Identification of two novel GATA6 mutations in an adult with acute myocardial infarction, diabetes, and atrial fibrillation: a case report. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2019; 16:785-788. [PMID: 31700519 PMCID: PMC6828608 DOI: 10.11909/j.issn.1671-5411.2019.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
37
|
Sun Z, Pang S, Cui Y, Yan B. Genetic and Functional Variants Analysis of the GATA6 Gene Promoter in Acute Myocardial Infarction. Front Genet 2019; 10:1100. [PMID: 31781165 PMCID: PMC6851265 DOI: 10.3389/fgene.2019.01100] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 10/11/2019] [Indexed: 01/13/2023] Open
Abstract
Background: Acute myocardial infarction (AMI) which is a specific type of coronary artery disease (CAD), is caused by the combination of genetic factors and acquired environment. Although some common genetic variations have been recorded to contribute to the development of CAD and AMI, more genetic factors and potential molecular mechanisms remain largely unknown. The GATA6 gene is expressed in the heart during embryogenesis and is also detected in vascular smooth muscle cells (VSMCs), different human primary endothelial cells (ECs), and vascular ECs in mice. To date, no studies have directly linked GATA6 gene with regulation of the CAD. Methods: In this study, we used a case-control study to investigate and analyze the genetic variations and functional variations of the GATA6 gene promoter region in AMI patients and controls. A variety of statistical analysis methods were utilized to analyze the association of single nucleotide polymorphisms (SNPs) with AMI. Functional analysis of DNA sequence variants (DSVs) was performed using a dual luciferase reporter assay. In vitro, electrophoretic mobility shift assay (EMSA) was selected to examine DNA-protein interactions. Results: A total of 705 subjects were enrolled in the study. Ten DSVs were found in AMI patients (n = 352) and controls (n = 353), including seven SNPs. One novel heterozygous DSV, (g.22168409 A > G), and two SNPs, [g.22168362 C > A(rs1416421760) and g.22168521 G > T(rs1445501474)], were reported in three AMI patients, which were not found in controls. The relevant statistical analysis, including allele and genotype frequencies between AMI patients and controls, five genetic models, linkage disequilibrium (LD) and haplotype analysis, and SNP–SNP interactions, suggested no statistical significance (P > 0.05). The transcriptional activity of GATA6 gene promoter was significantly increased by the DSV (g.22168409 A > G) and SNP [g.22168362 C > A(rs1416421760)]. The EMSA revealed that the DSV (g.22168409 A > G) and SNP [g.22168362 C > A(rs1416421760)] evidently influenced the binding of transcription factors. Conclusion: In conclusion, the DSV (g.22168409 A > G) and SNP [g.22168362 C > A(rs1416421760)] may increase GATA6 levels in both HEK-293 and H9c2 cell lines by affecting the binding of transcription factors. Whether the two variants identified in the GATA6 gene promoter can promote the development and progression of human AMI by altering GATA6 levels still requires further studies to verify.
Collapse
Affiliation(s)
- Zhaoqing Sun
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Yinghua Cui
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,The Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,The Center for Molecular Genetics of Cardiovascular Diseases, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China.,Shandong Provincial Sino-US Cooperation Research Center for Translational Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| |
Collapse
|
38
|
A genetic risk score predicts recurrent events after myocardial infarction in young adults. ACTA ACUST UNITED AC 2019; 73:623-631. [PMID: 31629691 DOI: 10.1016/j.rec.2019.08.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/05/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION AND OBJECTIVES To evaluate whether a genetic risk score (GRS) improves prediction of recurrent events in young nondiabetic patients presenting with an acute myocardial infarction (AMI) and identifies a more aggressive form of atherosclerosis. METHODS We conducted a prospective study with consecutive nondiabetic patients aged <55 years presenting with AMI. We performed a genetic test, cardiac computed tomography, and analyzed several biomarkers. We studied the association of a GRS composed of 11 genetic variants and a primary composite endpoint (cardiovascular mortality, a recurrent event, and cardiac hospitalization). RESULTS A total of 81 patients were studied and followed up for a median of 4.1 years. There were 24 recurrent cardiovascular events. Compared with the general population, study participants had a higher prevalence of 9 out of 11 risk alleles. The GRS was significantly associated with recurrent cardiovascular events, especially when baseline low-density lipoprotein cholesterol (LDL-C) levels were elevated. Compared with the low-risk GRS tertile, the multivariate-adjusted HR for recurrences was 10.2 (95%CI, 1.1-100.3; P=.04) for the intermediate-risk group and was 20.7 (2.4-181.0; P=.006) for the high-risk group when LDL-C was≥2.8mmol/L (≥ 110mg/dL). Inclusion of the GRS improved the C-statistic (ΔC-statistic=0.086), cNRI (continuous net reclassification improvement) (30%), and the IDI (integrated discrimination improvement) index (0.05). Cardiac computed tomography frequently detected coronary calcified atherosclerosis but had limited value for prediction of recurrences. No association was observed between metalloproteinases, GRS and recurrences. CONCLUSIONS A multilocus GRS may identify individuals at increased risk of long-term recurrences among young nondiabetic patients with AMI and improve clinical risk stratification models, particularly among patients with high baseline LDL-C levels.
Collapse
|
39
|
Prediction and management of CAD risk based on genetic stratification. Trends Cardiovasc Med 2019; 30:328-334. [PMID: 31543237 DOI: 10.1016/j.tcm.2019.08.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/01/2019] [Accepted: 08/20/2019] [Indexed: 12/24/2022]
Abstract
Discovery of genetic risk variants for CAD and their assembly on a computerized microarray enables a genetic risk score (GRS) to be expressed as a single number. Utilizing this array, genetic risk stratification has been performed in over 1 million cases and controls. The genetic score based on one's DNA can be determined anytime from birth on and is independent of age and conventional risk factors. Utilizing the GRS, one can select those at highest risk and would benefit most from primary prevention. Clinical trials have shown that modifying lifestyle or using statin therapy reduces the risk for CAD by approximately 50%. The use of the GRS for primary prevention will have a transformative effect on preventing the spread of CAD.
Collapse
|
40
|
Kobyakova OS, Kulikov ES, Malykh RD, Chernogoryuk GE, Deev IA, Starovoytova EA, Kirillova NA, Zagromova TA, Balaganskaya MA. Strategies for the prevention of chronic non-communicable diseases: a modern look at the problem. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2019. [DOI: 10.15829/1728-8800-2019-4-92-98] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
41
|
Santos-Ferreira C, Baptista R, Oliveira-Santos M, Costa R, Pereira Moura J, Gonçalves L. Apolipoprotein E2 Genotype Is Associated with a 2-Fold Increase in the Incidence of Type 2 Diabetes Mellitus: Results from a Long-Term Observational Study. J Lipids 2019; 2019:1698610. [PMID: 31485353 PMCID: PMC6702854 DOI: 10.1155/2019/1698610] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 07/11/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The apolipoprotein E (APOE) polymorphisms are associated with cardiovascular (CV) disease, but its interaction with type 2 diabetes mellitus (T2DM) long-term incidence is unknown. We investigated the association between APOE genotype and long-term (i) CV events and (ii) T2DM incidence in a Southern European primary prevention cohort. METHODS We assessed individual APOE genotypes in a total of 436 patients followed at a lipid clinic, with a 15-year median follow-up time. We collected data on major CV events (CV death, myocardial infarction, and stroke) and T2DM development. RESULTS No differences were found regarding major CV event incidence among the different APOE genotypes. However, after excluding 39 patients with a prior history of T2DM, APOE2 carriers displayed a higher incidence of T2DM during follow-up (42.2%) than APOE3 (27.1%) and APOE4 (28.7%) carriers. The age-, sex-, triglycerides-, and statin usage-adjusted OR for T2DM incidence in APOE2 carriers was 1.8 (95%CI 1.1-2.9, p=0.03), compared with wild-type APOE3. To address the role of statins as a confounder, we analyzed T2DM incidence in statin-treated patients. Statin-treated APOE2 carriers also had a higher T2DM incidence (57.9%), in comparison with APOE3 homozygotes (31.6%) and APOE4 carriers (32.5%). After adjustment for confounding, APOE2 carriers on statins displayed a similar twofold increase in T2DM risk compared to APOE3 homozygotes (OR 2.1, 95%CI 1.1-4.0, p=0.03). CONCLUSION Our findings suggest a twofold increase in T2DM incidence in APOE2 carriers. This may prompt for a specific glucose dysmetabolism follow-up that might be tailored on the APOE genotype.
Collapse
Affiliation(s)
| | - Rui Baptista
- Department of Cardiology, Coimbra University Hospital Center, Coimbra, Portugal
- iCBR, Faculty of Medicine, University of Coimbra, Portugal
| | | | - Regina Costa
- Department of Internal Medicine, Coimbra University Hospital Center, Coimbra, Portugal
| | - José Pereira Moura
- Department of Internal Medicine, Coimbra University Hospital Center, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Portugal
| | - Lino Gonçalves
- Department of Cardiology, Coimbra University Hospital Center, Coimbra, Portugal
- iCBR, Faculty of Medicine, University of Coimbra, Portugal
| |
Collapse
|
42
|
The Role of Genetics in Cardiovascular Risk Reduction: Findings From a Single Lipid Clinic and Review of the Literature. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2019; 21:200-204. [PMID: 31153847 DOI: 10.1016/j.carrev.2019.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/04/2019] [Accepted: 04/04/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND Genetic information is not routinely obtained in the management of most lipid disorders or in primary or secondary prevention of cardiovascular disease (CVD). We sought to determine the prevalence of pathogenic variants associated with lipoprotein metabolism or coronary artery disease (CAD) in a single lipid clinic and discuss the future use of genetic information in CVD prevention. METHODS Genetic testing was offered to patients with hypertriglyceridemia (defined as pre-treatment fasting triglycerides ≥150 mg/dL), elevated LDL-C (defined as pre-treatment ≥190 mg/dL), low HDL-C (defined as ≤40 mg/dL), elevated lipoprotein (a) (defined as ≥50 mg/dL or 100 nmol/L) or premature CAD (defined as an acute coronary syndrome or revascularization before age 40 years in men and 50 years in women) using next-generation DNA sequencing of 327 exons and selected variants in 129 genes known or suspected to be associated with lipoprotein metabolism or CAD. RESULTS 82 of 84 patients (97.6%) were found to have a variant associated with abnormal lipid metabolism or CAD. The most common pathogenic or likely pathogenic variants included those of the LDL receptor (15 patients) and lipoprotein lipase (9 patients). Other common variants included those of apolipoprotein A5 (14 patients) and variants associated with elevated lipoprotein (a) (25 patients). CONCLUSIONS The majority of patients presenting to a single lipid clinic were found to have at least one variant associated with abnormal lipoprotein metabolism or CAD. Incorporating genetic information, including the use of genetic risk scores, is anticipated in the future care of lipid disorders and CVD prevention.
Collapse
|
43
|
2017 Roadmap for Innovation-ACC Health Policy Statement on Healthcare Transformation in the Era of Digital Health, Big Data, and Precision Health: A Report of the American College of Cardiology Task Force on Health Policy Statements and Systems of Care. J Am Coll Cardiol 2019; 70:2696-2718. [PMID: 29169478 DOI: 10.1016/j.jacc.2017.10.018] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
44
|
Chen J, Wang S, Pang S, Cui Y, Yan B, Hawley RG. Functional genetic variants of the GATA4 gene promoter in acute myocardial infarction. Mol Med Rep 2019; 19:2861-2868. [PMID: 30720078 DOI: 10.3892/mmr.2019.9914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 01/25/2019] [Indexed: 11/05/2022] Open
Abstract
Coronary artery disease (CAD), including acute myocardial infarction (AMI), is a common complex disease; however, the genetic causes remain largely unknown. Recent epidemiological investigations indicated that the incidence of CAD in patients with congenital heart diseases is markedly higher than that observed in healthy controls. It was therefore hypothesized that the dysregulated expression of cardiac developmental genes may be involved in CAD development. GATA binding protein 4 (GATA4) serves essential roles in heart development and coronary vessel formation. In the present study, the GATA4 gene promoter was analyzed in patients with AMI (n=395) and in ethnically‑matched healthy controls (n=397). A total of 14 DNA variants were identified, including two single‑nucleotide polymorphisms. Three novel heterozygous DNA variants (g.31806C>T, g.31900G>C and g.32241C>T) were reported in three patients with AMI. These DNA variants significantly increased the activity of the GATA4 gene promoter. The electrophoretic mobility shift assay revealed that the DNA variant g.32241C>T influenced the binding ability of transcription factors. Taken together, the DNA variants may alter GATA4 gene promoter activity and affect GATA4 levels, thus contributing to AMI development.
Collapse
Affiliation(s)
- Jing Chen
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Shuai Wang
- Department of Medicine, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Yinghua Cui
- Division of Cardiology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and Treatment, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Robert G Hawley
- Department of Anatomy and Regenerative Biology, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
45
|
Sarraju A, Knowles JW. Genetic Testing and Risk Scores: Impact on Familial Hypercholesterolemia. Front Cardiovasc Med 2019; 6:5. [PMID: 30761309 PMCID: PMC6361766 DOI: 10.3389/fcvm.2019.00005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/11/2019] [Indexed: 12/11/2022] Open
Abstract
Familial Hypercholesterolemia (FH) is an inherited lipid disorder affecting 1 in 220 individuals resulting in highly elevated low-density lipoprotein levels and risk of premature coronary disease. Pathogenic variants causing FH typically involve the LDL receptor (LDLR), apolipoprotein B-100 (APOB), and proprotein convertase subtulisin/kexin type 9 genes (PCSK9) and if identified convey a risk of early onset coronary artery disease (ASCVD) of 3- to 10-fold vs. the general population depending on the severity of the mutation. Identification of monogenic FH within a family has implications for family-based testing (cascade screening), risk stratification, and potentially management, and it has now been recommended that such testing be offered to all potential FH patients. Recently, robust genome wide association studies (GWAS) have led to the recognition that the accumulation of common, small effect alleles affecting many LDL-c raising genes can result in a clinical phenotype largely indistinguishable from monogenic FH (i.e., a risk of early onset ASCVD of ~3-fold) in those at the extreme tail of the distribution for these alleles (i.e., the top 8% of the population for a polygenic risk score). The incorporation of these genetic risk scores into clinical practice for non-FH patients may improve risk stratification but is not yet widely performed due to a less robust evidence base for utility. Here, we review the current status of FH genetic testing, potential future applications as well as challenges and pitfalls.
Collapse
Affiliation(s)
- Ashish Sarraju
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Joshua W Knowles
- Division of Cardiovascular Medicine and Cardiovascular Institute, Stanford University, Stanford, CA, United States.,The FH Foundation, Pasadena, CA, United States.,Stanford Diabetes Research Center, Stanford University, Stanford, CA, United States
| |
Collapse
|
46
|
|
47
|
Matsuura Y, Kanter JE, Bornfeldt KE. Highlighting Residual Atherosclerotic Cardiovascular Disease Risk. Arterioscler Thromb Vasc Biol 2019; 39:e1-e9. [PMID: 30586334 PMCID: PMC6310032 DOI: 10.1161/atvbaha.118.311999] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yunosuke Matsuura
- From the Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle (Y.M., J.E.K., K.E.B.)
| | - Jenny E Kanter
- From the Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle (Y.M., J.E.K., K.E.B.)
| | - Karin E Bornfeldt
- From the Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle (Y.M., J.E.K., K.E.B.)
- Department of Pathology, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle (K.E.B.)
| |
Collapse
|
48
|
Ortendahl JD, Diamant AL, Toth PP, Cherepanov D, Harmon AL, Broder MS. Protecting the gains: What changes are needed to prevent a reversal of the downward cardiovascular disease mortality trend? Clin Cardiol 2018; 42:47-55. [PMID: 30318600 DOI: 10.1002/clc.23097] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 01/02/2023] Open
Abstract
AIMS Cardiovascular disease (CVD) mortality has decreased over 60% over the past 50 years in the United States; however, emerging data indicate CVD incidence may be rising because of shifting demographics, increasing risk factor prevalence, and competing needs for limited resources. We projected CVD mortality from 2015 to 2040 given varying informed assumptions regarding changes in risk factor prevalence, uptake of current therapeutic options, and future innovations. METHODS A microsimulation model was used to project US CVD mortality trends. National Health and Nutrition Examination Survey data were used to estimate population-level trends in CVD risk factors. Risk factors were used to generate Framingham Risk Scores for cohorts of 1 000 000 individuals from the general population to determine each individuals' CVD risk. Annual cardiovascular incidence, prevalence, and mortality were projected for scenarios differing by uptake of current therapies, anticipated pharmaceutical innovations with variable efficacy, risk factor prevalence, and changes in health disparities. RESULTS When incorporating a demographic shift, continued changes in risk factors, current treatment utilization, and no major innovations, we predicted the CVD mortality rate would increase 41% by 2040. If innovations providing incremental benefits equal to those associated with the introduction of statins are identified and widely utilized, CVD mortality could remain constant through 2040. With more efficacious innovations, CVD mortality could be further reduced. CONCLUSIONS Given demographic and risk prevalence changes, increasing access and adherence to current preventative therapeutics could slow the expected mortality increase, but new therapies may be needed to maintain the downward trend in CVD deaths.
Collapse
Affiliation(s)
- Jesse D Ortendahl
- Partnership for Health Analytic Research, LLC, Beverly Hills, California
| | - Allison L Diamant
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Peter P Toth
- Preventative Cardiology, CGH Medical Center, Sterling, Illinois.,Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dasha Cherepanov
- Partnership for Health Analytic Research, LLC, Beverly Hills, California
| | - Amanda L Harmon
- Partnership for Health Analytic Research, LLC, Beverly Hills, California
| | - Michael S Broder
- Partnership for Health Analytic Research, LLC, Beverly Hills, California
| |
Collapse
|
49
|
Roberts R. Mendelian Randomization Studies Promise to Shorten the Journey to FDA Approval. JACC Basic Transl Sci 2018; 3:690-703. [PMID: 30456340 PMCID: PMC6234613 DOI: 10.1016/j.jacbts.2018.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/19/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022]
Abstract
There has been a dearth of new drugs approved for cardiovascular disorders. The cost is prohibitive, averaging to $2.5 billion, and requiring 12.5 years. This is in large part due to the high failure rate, with only 5% approval by the Food and Drug Administration. Despite preclinical studies showing potential safety and efficacy, most fail when they go to clinical trials phase I to III. One cause for failure is the drug target, often discovered to be a biomarker rather than causative for the disease. Mendelian randomization (MR) studies would determine whether the drug target is causative and could save millions of dollars and time, and prevent unnecessary exposure to adverse drug effects. This was demonstrated in 3 clinical trials that were negative with 2 drugs, veraspladib and darapladib. MR studies during the trials showed the targets of secretory and lipoprotein-associated phospholipids A2 are not causative for coronary artery disease and predicted negative results. The requirement for MR studies is a genetic risk variant with altered function, randomized at conception that remains fixed throughout one’s lifetime. It is not confounded by dietary, lifestyle, or socioeconomic factors. It is more sensitive than randomized controlled trials because exposure to the risk factor is fixed for a lifetime. MR studies showed plasma high-density lipoprotein cholesterol is not a causative target of coronary artery disease, and neither is uric acid, C-reactive protein, and others. MR studies are highly sensitive in determining whether drug targets are causative, and are relatively easy, inexpensive, and not time consuming. It is recommended that drug targets undergo MR studies before proceeding to randomized controlled trials.
Collapse
Affiliation(s)
- Robert Roberts
- Department of Medicine, University of Arizona College of Medicine, Phoenix, Arizona
| |
Collapse
|
50
|
Zhang P, Zhang J, Zhang Y, Wang S, Pang S, Yan B. Functional variants of the ATG7 gene promoter in acute myocardial infarction. Mol Genet Genomic Med 2018; 6:1209-1219. [PMID: 30407747 PMCID: PMC6305654 DOI: 10.1002/mgg3.508] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 10/07/2018] [Accepted: 10/16/2018] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Coronary artery disease including acute myocardial infarction (AMI) is mainly caused by atherosclerosis, an inflammatory and metabolic disease. Autophagy has been demonstrated to play critical roles in lipid metabolism and inflammation. Altered autophagic activity has been reported in AMI patients. However, molecular basis for dysfunctional autophagy in AMI remains unexplained. METHODS In this study, the promoter of the ATG7 gene, encoding a core protein for autophagy, was genetically and functionally analyzed in large cohorts of AMI patients (n = 355) and ethnic-matched healthy controls (n = 363). Related molecular mechanisms were also explored. RESULTS A total of 19 DNA sequence variants (DSVs) including single-nucleotide polymorphisms (SNPs) were found in the ATG7 gene promoter. Two novel DSVs and five SNPs were only identified in AMI patients group. These DSVs and SNPs, except one SNP, significantly altered the transcriptional activity of the ATG7 gene promoter in both HEK-293 and H9c2 cells (p < 0.05). Further electrophoretic mobility shift assay revealed that the DSVs and SNPs evidently affected the binding of transcription factors. CONCLUSIONS ATG7 gene DSVs and SNPs identified in AMI patients may alter the transcriptional activity of the ATG7 gene promoter and change ATG7 level, contributing to the AMI development as a rare risk factor.
Collapse
Affiliation(s)
- Pei Zhang
- College of Clinical MedicineXinxiang Medical UniversityXinxiangHenanChina
- Division of EmergencyJining First People's HospitalJiningShandongChina
| | - Jie Zhang
- Department of MedicineShandong University School of MedicineJinanShandongChina
| | - Yexin Zhang
- Department of MedicineShandong University School of MedicineJinanShandongChina
| | - Shuai Wang
- Department of MedicineShandong University School of MedicineJinanShandongChina
| | - Shuchao Pang
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and TreatmentAffiliated Hospital of Jining Medical UniversityJining Medical UniversityJiningShandongChina
| | - Bo Yan
- Shandong Provincial Key Laboratory of Cardiac Disease Diagnosis and TreatmentAffiliated Hospital of Jining Medical UniversityJining Medical UniversityJiningShandongChina
- The Center for Molecular Genetics of Cardiovascular DiseasesAffiliated Hospital of Jining Medical UniversityJining Medical UniversityJiningShandongChina
- Shandong Provincial Sino‐US Cooperation Research Center for Translational MedicineAffiliated Hospital of Jining Medical UniversityJining Medical UniversityJiningShandongChina
| |
Collapse
|