1
|
Aminbakhsh AP, Théberge ET, Burden E, Adejumo CK, Gravely AK, Lehman A, Sedlak TL. Exploring associations between estrogen and gene candidates identified by coronary artery disease genome-wide association studies. Front Cardiovasc Med 2025; 12:1502985. [PMID: 40182431 PMCID: PMC11965610 DOI: 10.3389/fcvm.2025.1502985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/04/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Coronary artery disease (CAD) is the leading cause of death around the world, with epidemiological sex and gender differences in prevalence, pathophysiology and outcomes. It has been hypothesized that sex steroids, like estrogen, may contribute to these sex differences. There is a relatively large genetic component to developing CAD, with heritability estimates ranging between 40%-60%. In the last two decades, genome-wide association studies (GWAS) have contributed substantially to advancing the understanding of genetic candidates contributing to CAD. The aim of this study was to determine if genes discovered in CAD GWASs are affected by estrogen via direct modulation or indirect down-stream targets. Methods A scoping review was conducted using MEDLINE and EMBASE for studies of atherosclerotic coronary artery disease and a genome-wide association study (GWAS) design. Analysis was limited to candidate genes with corresponding single nucleotide polymorphisms (SNPs) surpassing genome-wide significance and had been mapped to genes by study authors. The number of studies that conducted sex-stratified analyses with significant genes were quantified. A literature search of the final gene lists was done to examine any evidence suggesting estrogen may modulate the genes and/or gene products. Results There were 60 eligible CAD GWASs meeting inclusion criteria for data extraction. Of these 60, only 36 had genome-wide significant SNPs reported, and only 3 of these had significant SNPs from sex-stratified analyses mapped to genes. From these 36 studies, a total of 61 genes were curated, of which 26 genes (43%) were found to have modulation by estrogen. All 26 were discovered in studies that adjusted for sex. 12/26 genes were also discovered in studies that conducted sex-stratified analyses. 12/26 genes were classified as having a role in lipid synthesis, metabolism and/or lipoprotein mechanisms, while 11/26 were classified as having a role in vascular integrity, and 3/26 were classified as having a role in thrombosis. Discussion This study provides further evidence of the relationship between estrogen, genetic risk and the development of CAD. More sex-stratified research will need to be conducted to further characterize estrogen's relation to sex differences in the pathology and progression of CAD.
Collapse
Affiliation(s)
- Ava P. Aminbakhsh
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Emilie T. Théberge
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth Burden
- Division of Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Cindy Kalenga Adejumo
- Division of Internal Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Annabel K. Gravely
- Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Anna Lehman
- Department of Medical Genetics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Vancouver Coastal Health, Vancouver, BC, Canada
| | - Tara L. Sedlak
- Vancouver Coastal Health, Vancouver, BC, Canada
- Division of Cardiology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
2
|
Popov VS, Brodsky IB, Balatskaya MN, Balatskiy AV, Ozhimalov ID, Kulebyakina MA, Semina EV, Arbatskiy MS, Isakova VS, Klimovich PS, Sysoeva VY, Kalinina NI, Tkachuk VA, Rubina KA. T-Cadherin Deficiency Is Associated with Increased Blood Pressure after Physical Activity. Int J Mol Sci 2023; 24:14204. [PMID: 37762507 PMCID: PMC10531645 DOI: 10.3390/ijms241814204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/07/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
T-cadherin is a regulator of blood vessel remodeling and angiogenesis, involved in adiponectin-mediated protective effects in the cardiovascular system and in skeletal muscles. GWAS study has previously demonstrated a SNP in the Cdh13 gene to be associated with hypertension. However, the role of T-cadherin in regulating blood pressure has not been experimentally elucidated. Herein, we generated Cdh13∆Exon3 mice lacking exon 3 in the Cdh13 gene and described their phenotype. Cdh13∆Exon3 mice exhibited normal gross morphology, life expectancy, and breeding capacity. Meanwhile, their body weight was considerably lower than of WT mice. When running on a treadmill, the time spent running and the distance covered by Cdh13∆Exon3 mice was similar to that of WT. The resting blood pressure in Cdh13∆Exon3 mice was slightly higher than in WT, however, upon intensive physical training their systolic blood pressure was significantly elevated. While adiponectin content in the myocardium of Cdh13∆Exon3 and WT mice was within the same range, adiponectin plasma level was 4.37-fold higher in Cdh13∆Exon3 mice. Moreover, intensive physical training augmented the AMPK phosphorylation in the skeletal muscles and myocardium of Cdh13∆Exon3 mice as compared to WT. Our data highlight a critically important role of T-cadherin in regulation of blood pressure and stamina in mice, and may shed light on the pathogenesis of hypertension.
Collapse
Affiliation(s)
- Vladimir S. Popov
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Ilya B. Brodsky
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Maria N. Balatskaya
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Alexander V. Balatskiy
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Ilia D. Ozhimalov
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Maria A. Kulebyakina
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Ekaterina V. Semina
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Mikhail S. Arbatskiy
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Viktoria S. Isakova
- V.I. Kulakov National Medical Center of Obstetrics Gynecology and Perinatology, Akademika Oparina Street, 4, 117198 Moscow, Russia
| | - Polina S. Klimovich
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Veronika Y. Sysoeva
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Natalia I. Kalinina
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Vsevolod A. Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| | - Kseniya A. Rubina
- Faculty of Medicine, Lomonosov Moscow State University, Lomonosovsky Ave., 27/1, 119991 Moscow, Russia (I.B.B.); (M.N.B.)
| |
Collapse
|
3
|
Clark KC, Kwitek AE. Multi-Omic Approaches to Identify Genetic Factors in Metabolic Syndrome. Compr Physiol 2021; 12:3045-3084. [PMID: 34964118 PMCID: PMC9373910 DOI: 10.1002/cphy.c210010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Metabolic syndrome (MetS) is a highly heritable disease and a major public health burden worldwide. MetS diagnosis criteria are met by the simultaneous presence of any three of the following: high triglycerides, low HDL/high LDL cholesterol, insulin resistance, hypertension, and central obesity. These diseases act synergistically in people suffering from MetS and dramatically increase risk of morbidity and mortality due to stroke and cardiovascular disease, as well as certain cancers. Each of these component features is itself a complex disease, as is MetS. As a genetically complex disease, genetic risk factors for MetS are numerous, but not very powerful individually, often requiring specific environmental stressors for the disease to manifest. When taken together, all sequence variants that contribute to MetS disease risk explain only a fraction of the heritable variance, suggesting additional, novel loci have yet to be discovered. In this article, we will give a brief overview on the genetic concepts needed to interpret genome-wide association studies (GWAS) and quantitative trait locus (QTL) data, summarize the state of the field of MetS physiological genomics, and to introduce tools and resources that can be used by the physiologist to integrate genomics into their own research on MetS and any of its component features. There is a wealth of phenotypic and molecular data in animal models and humans that can be leveraged as outlined in this article. Integrating these multi-omic QTL data for complex diseases such as MetS provides a means to unravel the pathways and mechanisms leading to complex disease and promise for novel treatments. © 2022 American Physiological Society. Compr Physiol 12:1-40, 2022.
Collapse
Affiliation(s)
- Karen C Clark
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Anne E Kwitek
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
4
|
Rubina KA, Semina EV, Kalinina NI, Sysoeva VY, Balatskiy AV, Tkachuk VA. Revisiting the multiple roles of T-cadherin in health and disease. Eur J Cell Biol 2021; 100:151183. [PMID: 34798557 DOI: 10.1016/j.ejcb.2021.151183] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 01/02/2023] Open
Abstract
As a non-canonical member of cadherin superfamily, T-cadherin was initially described as a molecule involved in homophilic recognition in the nervous and vascular systems. The ensuing decades clearly demonstrated that T-cadherin is a remarkably multifunctional molecule. It was validated as a bona fide receptor for both: LDL exerting adverse atherogenic action and adiponectin mediating many protective metabolic and cardiovascular effects. Motivated by the latest progress and accumulated data unmasking important roles of T-cadherin in blood vessel function and tissue regeneration, here we revisit the original function of T-cadherin as a guidance receptor for the growing axons and blood vessels, consider the recent data on T-cadherin-induced exosomes' biogenesis and their role in myocardial regeneration and revascularization. The review expands upon T-cadherin contribution to mesenchymal stem/stromal cell compartment in adipose tissue. We also dwell upon T-cadherin polymorphisms (SNP) and their possible therapeutic applications. Furthermore, we scrutinize the molecular hub of insulin and adiponectin receptors (AdipoR1 and AdipoR2) conveying signals to their downstream targets in quest for defining a putative place of T-cadherin in this molecular circuitry.
Collapse
Affiliation(s)
- K A Rubina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia.
| | - E V Semina
- Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - N I Kalinina
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - V Yu Sysoeva
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - A V Balatskiy
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia
| | - V A Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, 119192 Moscow, Russia; Institute of Experimental Cardiology, National Cardiology Research Center of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| |
Collapse
|
5
|
Bojang KS, Lyrawati D, Sujuti H, Wahono D. Association of CDH13 Gene Polymorphism and Metabolic Syndrome in Gambian Population. Med Arch 2021; 75:262-268. [PMID: 34759445 PMCID: PMC8563038 DOI: 10.5455/medarh.2021.75.262-268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/10/2021] [Indexed: 11/06/2022] Open
Abstract
Background: Polymorphism in CDH13 gene, which encodes for the adiponectin receptor, T-cadherin, is a genetic risk factor associated with metabolic syndrome. CDH13 rs3865188, which is found in the promoter region of the CDH13 gene, has been found to be associated with metabolic syndrome and its traits in Asian and European Caucasian populations. However, to the best of our knowledge, it was yet to be assessed in a Black African population. Objective: The aim of this study was to investigate the association of CHD13 rs3865188 and metabolic syndrome in a Gambian population. Methods: It was a genetic association study in a cross-sectional design in 136 Gambian participants. CDH13 rs3865188 was genotyped using PCR master mix and sequencing. Blood sugar, triglyceride and high-density lipoprotein levels were determined by standard clinical laboratory methods. Results: CDH13 rs3865188 was found to be significantly associated metabolic syndrome (p=0.034). Genotype AT appeared to be risk factor for metabolic syndrome (OR=2.41, 95% CI, 1.20–4.84, p=0.014). We found genotypes CC and CA in CHD13 rs3865188 for the first time. Conclusion: Our study demonstrated significant association between CDH13 rs385618 and metabolic syndrome in a Gambian population (Black African population for the first time). Individuals with genotype AT are at higher risk of developing metabolic syndrome.
Collapse
Affiliation(s)
- Kebba S Bojang
- Doctoral Program in Medical Science, Faculty of Medicine, Brawijaya University, Malang, Indonesia.,School of Medicine and Allied Health Sciences, University of The Gambia, Banjul, The Gambia.,Department of Internal Medicine, Kanifing General Hospital, Kanifing, The Gambia
| | - Diana Lyrawati
- Doctoral Program in Medical Science, Faculty of Medicine, Brawijaya University, Malang, Indonesia.,Department of Pharmacy, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Hidayat Sujuti
- Doctoral Program in Medical Science, Faculty of Medicine, Brawijaya University, Malang, Indonesia.,Department of Ophthalmology, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Djoko Wahono
- Doctoral Program in Medical Science, Faculty of Medicine, Brawijaya University, Malang, Indonesia.,Department of Internal Medicine, Faculty of Medical, Brawijaya University, Malang, Indonesia
| |
Collapse
|
6
|
Genome-Wide Association Study on Adiponectin-Mediated Suppression of HDL-C Levels in Taiwanese Individuals Identifies Functional Haplotypes in CDH13. Genes (Basel) 2021; 12:genes12101582. [PMID: 34680977 PMCID: PMC8535967 DOI: 10.3390/genes12101582] [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: 09/02/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/24/2022] Open
Abstract
CDH13 encodes T-cadherin, which is expressed in the vasculature and cardiac myocytes and is the receptor for hexameric and high-molecular-weight adiponectin. The CDH13 region is the most pivotal locus associated with adiponectin level. Mediation analysis is a method to explore the effect of a third variable, it is assumed that the magnitude of the relationship between the independent and dependent variables will be reduced by statistical adjustment for a third variable. In addition, mediation can further occur in the case when the mediator acts as a pathway-suppressor variable that means a suppression effect may be suggested if the statistical removal of a mediation effect could increase the magnitude of the relationship between the independent and dependent variables. Here, we aimed to explore the suppression effect in a genome-wide association study, and investigate possible mechanisms that may link adiponectin to CDH13 variants and high-density lipoprotein cholesterol (HDL-C). Genome-wide association data for adiponectin and HDL-C were accessible for 2349 Taiwan-biobank participants. The mediation analysis was conducted with the CDH13 lead single nucleotide polymorphism (SNP) rs4783244. The cloned constructs of CDH13 haplotypes (GG and TT) identified from the rs4783244 G/T and rs12051272 G/T SNPs were transiently expressed in HEK293T cells and investigated using the luciferase reporter assay. Genome-wide association analysis showed that HDL-C is significantly associated with variants in CDH13 after adjusting for the adiponectin level. The lead SNP rs4783244 was significantly associated with lower adiponectin levels and exhibited a suppression effect on HDL-C when adiponectin was included as a third factor in the mediation analysis. Luciferase reporter assay results further demonstrated that the GG haplotype increased enhancer activity, whereas the haplotype TT significantly reduced the activity of this enhancer. We present the first evidence of the suppressive role of adiponectin in the genome-wide association between CDH13 and HDL-C. CDH13 may increase the HDL-C levels, and its expression is suppressed by adiponectin.
Collapse
|
7
|
T-Cadherin and the Ratio of Its Ligands as Predictors of Carotid Atherosclerosis: A Pilot Study. Biomedicines 2021; 9:biomedicines9101398. [PMID: 34680515 PMCID: PMC8533356 DOI: 10.3390/biomedicines9101398] [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: 09/01/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 12/27/2022] Open
Abstract
In the cardiovascular system, atherogenic low-density lipoproteins (LDL) and the protective hormone adiponectin bind to the same receptor, T-cadherin. In this study, we tested the hypothesis that the ratio of circulating LDL to high-molecular weight (HMW) adiponectin could predict the development of atherosclerosis. Using enzyme-linked immunosorbent assay, we measured the level of circulating HMW adiponectin in the blood of donors together with ultrasound measuring of intima-media thickness (IMT) of carotid arteries. Single-nucleotide polymorphisms in the T-cadherin gene were identified using polymerase chain reaction. We found that carotid artery IMT is inversely correlated with the level of HMW in male subjects. We also found that the G allele of rs12444338 SNP in the T-cadherin gene correlates with a lower level of circulating T-cadherin and thinner IMT and therefore could be considered as an atheroprotective genotype. Despite our data, we could not provide direct evidence for the initial study hypothesis. However, we did uncover an important correlation between circulating T-cadherin and thinner carotid IMT.
Collapse
|
8
|
McCaffrey TA, Toma I, Yang Z, Katz R, Reiner J, Mazhari R, Shah P, Tackett M, Jones D, Jepson T, Falk Z, Wargodsky R, Shtakalo D, Antonets D, Ertle J, Kim JH, Lai Y, Arslan Z, Aledort E, Alfaraidy M, Laurent GS. RNA sequencing of blood in coronary artery disease: involvement of regulatory T cell imbalance. BMC Med Genomics 2021; 14:216. [PMID: 34479557 PMCID: PMC8414682 DOI: 10.1186/s12920-021-01062-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 08/19/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Cardiovascular disease had a global prevalence of 523 million cases and 18.6 million deaths in 2019. The current standard for diagnosing coronary artery disease (CAD) is coronary angiography. Surprisingly, despite well-established clinical indications, up to 40% of the one million invasive cardiac catheterizations return a result of 'no blockage'. The present studies employed RNA sequencing of whole blood to identify an RNA signature in patients with angiographically confirmed CAD. METHODS Whole blood RNA was depleted of ribosomal RNA (rRNA) and analyzed by single-molecule sequencing of RNA (RNAseq) to identify transcripts associated with CAD (TRACs) in a discovery group of 96 patients presenting for elective coronary catheterization. The resulting transcript counts were compared between groups to identify differentially expressed genes (DEGs). RESULTS Surprisingly, 98% of DEGs/TRACs were down-regulated ~ 1.7-fold in patients with mild to severe CAD (> 20% stenosis). The TRACs were independent of comorbid risk factors for CAD, such as sex, hypertension, and smoking. Bioinformatic analysis identified an enrichment in transcripts such as FoxP1, ICOSLG, IKZF4/Eos, SMYD3, TRIM28, and TCF3/E2A that are likely markers of regulatory T cells (Treg), consistent with known reductions in Tregs in CAD. A validation cohort of 80 patients confirmed the overall pattern (92% down-regulation) and supported many of the Treg-related changes. TRACs were enriched for transcripts associated with stress granules, which sequester RNAs, and ciliary and synaptic transcripts, possibly consistent with changes in the immune synapse of developing T cells. CONCLUSIONS These studies identify a novel mRNA signature of a Treg-like defect in CAD patients and provides a blueprint for a diagnostic test for CAD. The pattern of changes is consistent with stress-related changes in the maturation of T and Treg cells, possibly due to changes in the immune synapse.
Collapse
Affiliation(s)
- Timothy A McCaffrey
- Division of Genomic Medicine, Department of Medicine, The George Washington Medical Center, The George Washington University, 2300 I Street NW, Ross Hall 443A, Washington, DC, 20037, USA.
- The St. Laurent Institute, Vancouver, WA, USA.
- Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, 20037, USA.
- True Bearing Diagnostics, Washington, DC, 20037, USA.
| | - Ian Toma
- Division of Genomic Medicine, Department of Medicine, The George Washington Medical Center, The George Washington University, 2300 I Street NW, Ross Hall 443A, Washington, DC, 20037, USA
- Department of Clinical Research and Leadership, The George Washington University, Washington, DC, 20037, USA
- True Bearing Diagnostics, Washington, DC, 20037, USA
| | - Zhaoquing Yang
- Division of Genomic Medicine, Department of Medicine, The George Washington Medical Center, The George Washington University, 2300 I Street NW, Ross Hall 443A, Washington, DC, 20037, USA
| | - Richard Katz
- Division of Cardiology, Department of Medicine, The George Washington University , Washington, DC, 20037, USA
| | - Jonathan Reiner
- Division of Cardiology, Department of Medicine, The George Washington University , Washington, DC, 20037, USA
| | - Ramesh Mazhari
- Division of Cardiology, Department of Medicine, The George Washington University , Washington, DC, 20037, USA
| | - Palak Shah
- Inova Heart and Vascular Institute, Fairfax, VA, USA
| | | | | | - Tisha Jepson
- SeqLL, Inc., Woburn, MA, USA
- The St. Laurent Institute, Vancouver, WA, USA
- True Bearing Diagnostics, Washington, DC, 20037, USA
| | - Zachary Falk
- Division of Genomic Medicine, Department of Medicine, The George Washington Medical Center, The George Washington University, 2300 I Street NW, Ross Hall 443A, Washington, DC, 20037, USA
| | - Richard Wargodsky
- Division of Genomic Medicine, Department of Medicine, The George Washington Medical Center, The George Washington University, 2300 I Street NW, Ross Hall 443A, Washington, DC, 20037, USA
| | - Dmitry Shtakalo
- A.P. Ershov Institute of Informatics Systems SB RAS, 6, Acad. Lavrentjeva Ave, Novosibirsk, Russia, 630090
| | - Denis Antonets
- A.P. Ershov Institute of Informatics Systems SB RAS, 6, Acad. Lavrentjeva Ave, Novosibirsk, Russia, 630090
| | - Justin Ertle
- Division of Genomic Medicine, Department of Medicine, The George Washington Medical Center, The George Washington University, 2300 I Street NW, Ross Hall 443A, Washington, DC, 20037, USA
| | - Ju H Kim
- Division of Cardiology, Department of Medicine, The George Washington University , Washington, DC, 20037, USA
| | - Yinglei Lai
- Department of Statistics, Biostatistics Center, The George Washington University, Washington, DC, 20037, USA
| | - Zeynep Arslan
- Division of Genomic Medicine, Department of Medicine, The George Washington Medical Center, The George Washington University, 2300 I Street NW, Ross Hall 443A, Washington, DC, 20037, USA
| | - Emily Aledort
- Division of Genomic Medicine, Department of Medicine, The George Washington Medical Center, The George Washington University, 2300 I Street NW, Ross Hall 443A, Washington, DC, 20037, USA
| | - Maha Alfaraidy
- Division of Genomic Medicine, Department of Medicine, The George Washington Medical Center, The George Washington University, 2300 I Street NW, Ross Hall 443A, Washington, DC, 20037, USA
| | | |
Collapse
|
9
|
Litviňuková M, Talavera-López C, Maatz H, Reichart D, Worth CL, Lindberg EL, Kanda M, Polanski K, Heinig M, Lee M, Nadelmann ER, Roberts K, Tuck L, Fasouli ES, DeLaughter DM, McDonough B, Wakimoto H, Gorham JM, Samari S, Mahbubani KT, Saeb-Parsy K, Patone G, Boyle JJ, Zhang H, Zhang H, Viveiros A, Oudit GY, Bayraktar OA, Seidman JG, Seidman CE, Noseda M, Hubner N, Teichmann SA. Cells of the adult human heart. Nature 2020; 588:466-472. [PMID: 32971526 PMCID: PMC7681775 DOI: 10.1038/s41586-020-2797-4] [Citation(s) in RCA: 1008] [Impact Index Per Article: 201.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Advanced insights into disease mechanisms and therapeutic strategies require a deeper understanding of the molecular processes involved in the healthy heart. Knowledge of the full repertoire of cardiac cells and their gene expression profiles is a fundamental first step in this endeavour. Here, using state-of-the-art analyses of large-scale single-cell and single-nucleus transcriptomes, we characterize six anatomical adult heart regions. Our results highlight the cellular heterogeneity of cardiomyocytes, pericytes and fibroblasts, and reveal distinct atrial and ventricular subsets of cells with diverse developmental origins and specialized properties. We define the complexity of the cardiac vasculature and its changes along the arterio-venous axis. In the immune compartment, we identify cardiac-resident macrophages with inflammatory and protective transcriptional signatures. Furthermore, analyses of cell-to-cell interactions highlight different networks of macrophages, fibroblasts and cardiomyocytes between atria and ventricles that are distinct from those of skeletal muscle. Our human cardiac cell atlas improves our understanding of the human heart and provides a valuable reference for future studies.
Collapse
Affiliation(s)
- Monika Litviňuková
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Carlos Talavera-López
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,EMBL - EBI, Wellcome Genome Campus, Hinxton, UK
| | - Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Daniel Reichart
- Department of Genetics, Harvard Medical School, Boston, MA, USA.,Department of Cardiology, University Heart & Vascular Center, University of Hamburg, Hamburg, Germany
| | - Catherine L Worth
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Masatoshi Kanda
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Department of Rheumatology and Clinical Immunology, Sapporo Medical University, Sapporo, Japan
| | - Krzysztof Polanski
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Matthias Heinig
- Institute of Computational Biology (ICB), HMGU, Neuherberg, Germany.,Department of Informatics, Technische Universitaet Muenchen (TUM), Munich, Germany
| | - Michael Lee
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Kenny Roberts
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Liz Tuck
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Eirini S Fasouli
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | - Barbara McDonough
- Department of Genetics, Harvard Medical School, Boston, MA, USA.,Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA.,Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Joshua M Gorham
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Sara Samari
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Krishnaa T Mahbubani
- Department of Surgery, University of Cambridge, NIHR Cambridge Biomedical Centre, Cambridge Biorepository for Translational Medicine, Cambridge, UK
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, NIHR Cambridge Biomedical Centre, Cambridge Biorepository for Translational Medicine, Cambridge, UK
| | - Giannino Patone
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Joseph J Boyle
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Hongbo Zhang
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.,Department of Histology and Embryology of Zhongshan School of Medicine, Sun-Yat Sen University, Guangzhou, China
| | - Hao Zhang
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Anissa Viveiros
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.,Mazankowski Alberta Heart Institute, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Omer Ali Bayraktar
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - J G Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
| | - Christine E Seidman
- Department of Genetics, Harvard Medical School, Boston, MA, USA. .,Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA. .,Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Michela Noseda
- National Heart and Lung Institute, Imperial College London, London, UK. .,British Heart Foundation Centre of Regenerative Medicine, British Heart Foundation Centre of Research Excellence, Imperial College London, London, UK.
| | - Norbert Hubner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany. .,Charité-Universitätsmedizin, Berlin, Germany. .,Berlin Institute of Health (BIH), Berlin, Germany.
| | - Sarah A Teichmann
- Cellular Genetics Programme, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK. .,Deptartment of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK.
| |
Collapse
|
10
|
Deng W, Liu H, Luo S, Clarke J, Glass C, Su L, Lin L, Christiani DC, Wei Q. APOB Genotypes and CDH13 Haplotypes in the Cholesterol-Related Pathway Genes Predict Non-Small Cell Lung Cancer Survival. Cancer Epidemiol Biomarkers Prev 2020; 29:1204-1213. [PMID: 32238407 PMCID: PMC7269811 DOI: 10.1158/1055-9965.epi-19-1262] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 01/07/2020] [Accepted: 03/20/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Several oncogenic signals are involved in the synthesis, metabolism, transportation, and modulation of cholesterol. However, the roles of genetic variants of the cholesterol pathway genes in cancer survival remain unclear. METHODS We investigated associations between 26,781 common SNPs in 209 genes of the cholesterol pathway and non-small cell lung cancer (NSCLC) survival by utilizing genotyping data from two published genome-wide association studies. We used multivariate Cox proportional hazards regression and expression quantitative trait loci analyses to identify survival-associated SNPs and their correlations with the corresponding mRNA expression, respectively. We also used the Kaplan-Meier survival analysis and bioinformatics functional prediction to further evaluate the identified independent SNPs. RESULTS We found five independent SNPs (APOB rs1801701C>T; CDH13 rs35859010 C>T, rs1833970 T>A, rs254315 T>C, and rs425904 T>C) to be significantly associated with NSCLC survival in both discovery and replication datasets. When the unfavorable genotype (APOB rs1801701CC) and haplotypes (CDH13 rs35859010-rs1833970-rs254315-rs425904 C-A-T-C and T-T-T-T) were combined into a genetic score as the number of unfavorable genotypes/haplotypes (NUGH) in the multivariate analysis, an increased NUGH was associated with worse survival (P trend < 0.0001). In addition, both APOB rs1801701T CONCLUSIONS Genetic variants of APOB and CDH13 in the cholesterol pathway were associated with NSCLC survival, possibly by affecting their gene expression. IMPACT Genetic variants of APOB and CDH13 in the cholesterol pathway may provide new scientific insights into NSCLC prognosis.
Collapse
Affiliation(s)
- Wei Deng
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Hongliang Liu
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
| | - Sheng Luo
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - Jeffrey Clarke
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Carolyn Glass
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, China
- Department of Pathology, Duke University School of Medicine, Durham, North Carolina
| | - Li Su
- Departments of Environmental Health and Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Lijuan Lin
- Departments of Environmental Health and Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - David C Christiani
- Departments of Environmental Health and Epidemiology, Harvard School of Public Health, Boston, Massachusetts
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Qingyi Wei
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina.
- Department of Population Health Sciences, Duke University School of Medicine, Durham, North Carolina
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| |
Collapse
|
11
|
Peng XY, Wang Y, Hu H, Zhang XJ, Li Q. Identification of the molecular subgroups in coronary artery disease by gene expression profiles. J Cell Physiol 2019; 234:16540-16548. [PMID: 30805932 DOI: 10.1002/jcp.28324] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/24/2019] [Accepted: 01/28/2019] [Indexed: 01/24/2023]
Abstract
Coronary artery disease (CAD) is the most common type of cardiovascular disease and becomes a leading cause of death worldwide. Aiming to uncover the underlying molecular features for different types of CAD, we classified 352 CAD cases into three subgroups based on gene expression profiles, which were retrieved from the Gene Expression Omnibus database. Also, these subgroups present different expression patterns and clinical characteristics. To uncover the transcriptomic differences between the subgroups, weighted gene co-expression analysis (WGCNA) was used and identified six subgroup-specific WGCNA modules. Characterization of the WCGNA modules revealed that lipid metabolism pathways, specifically upregulated in subgroup I, might be an indicator of increased severity. Moreover, subgroup II was considered as an early-stage of CAD because of normal-like gene expression patterns. In contrast, the mammalian target of rapamycin signaling pathway was significantly upregulated in subgroup III. Although subgroups II and III did not have a significant prognostic difference, their intrinsic biological characteristics were highly different, suggesting that the transcriptome classification may represent risk factors of both age and the intrinsic biological characteristics. In conclusion, the transcriptome classification of CAD cases revealed that cases from different subgroups may have their unique gene expression patterns, indicating that patients in each subgroup should receive more personalized treatment.
Collapse
Affiliation(s)
- Xiao-Yan Peng
- Department of Neurology, First People's Hospital of Jingzhou, First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Yong Wang
- Cardiovascular Disease Center, Central Hospital of Enshi Autonomous Prefecture, Enshi Clinical College of Wuhan University, Enshi, China
| | - Haibo Hu
- Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Xian-Jin Zhang
- Department of Intensive Care Unit, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huaian, China
| | - Qi Li
- Department of Emergency, Huai'an Hospital, Huai'an, China
| |
Collapse
|
12
|
Vilne B, Schunkert H. Integrating Genes Affecting Coronary Artery Disease in Functional Networks by Multi-OMICs Approach. Front Cardiovasc Med 2018; 5:89. [PMID: 30065929 PMCID: PMC6056735 DOI: 10.3389/fcvm.2018.00089] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 06/22/2018] [Indexed: 12/26/2022] Open
Abstract
Coronary artery disease (CAD) and myocardial infarction (MI) remain among the leading causes of mortality worldwide, urgently demanding a better understanding of disease etiology, and more efficient therapeutic strategies. Genetic predisposition as well as the environment and lifestyle are thought to contribute to disease risk. It is likely that non-linear and complex interactions occur between these multiple factors, involving simultaneous pathological changes in diverse cell types, tissues, and organs, at multiple molecular levels. Recent technological advances have exponentially expanded the breadth of available -omics data, from genome, epigenome, transcriptome, proteome, metabolome to even the microbiome. Integration of multiple layers of information across several -omics domains, i.e., the so-called multi-omics approach, currently holds the promise as a path toward precision medicine. Indeed, a more meaningful interpretation of genotype-phenotype relationships and the development of successful therapeutics tailored to individual patients are urgently needed. In this review, we will summarize recent findings and applications of integrative multi-omics in elucidating the etiology of CAD/MI; with a special focus on established disease susceptibility loci sequentially identified in genome-wide association studies (GWAS) over the last 10 years. Moreover, in addition to the autosomal genome, we will also consider the genetic variation in our “second genome”—the mitochondrial genome. Finally, we will summarize the current challenges in the field and point to future research directions required in order to successfully and effectively apply these approaches for precision medicine.
Collapse
Affiliation(s)
- Baiba Vilne
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany.,Munich Heart Alliance, German Centre for Cardiovascular Research, Munich, Germany
| |
Collapse
|
13
|
Li G, Feng D, Wang Y, Fu J, Han L, Li L, Grant SFA, Li M, Li M, Gao S. Loss of Cardio-Protective Effects at the CDH13 Locus Due to Gene-Sleep Interaction: The BCAMS Study. EBioMedicine 2018; 32:164-171. [PMID: 29903569 PMCID: PMC6020797 DOI: 10.1016/j.ebiom.2018.05.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 12/17/2022] Open
Abstract
Left ventricular mass index (LVMI) provides a metric for cardiovascular disease risk. We aimed to assess the association of adiponectin-related genetic variants resulting from GWAS in East Asians (loci in/near CDH13, ADIPOQ, WDR11FGF, CMIP and PEPD) with LVMI, and to examine whether sleep duration modified these genetic associations in youth. The 559 subjects aged 15–28 years were recruited from the Beijing Child and Adolescent Metabolic Syndrome study. Among the six loci, CDH13 rs4783244 was significantly correlated with adiponectin levels (p = 8.07 × 10−7). The adiponectin-rising allele in rs4783244 locus was significantly associated with decreased LVMI (p = 6.99 × 10−4) after adjusting for classical cardiovascular risk factors, and further for adiponectin levels, while no significant association was found between the other loci and LVMI. Moreover, we observed a significant interaction effect between rs4783244 and sleep duration (p = .005) for LVMI; the genetic association was more evident in long sleep duration while lost in short sleep duration. Similar interaction was found in the subgroup analysis using longitudinal data (p = .025 for interaction). In this young Chinese population, CDH13 rs4783244 represents a key locus for cardiac structure, and confers stronger cardio-protection in longer sleep duration when contrasted with short sleep duration. CDH13 rs4783244 represents a key locus for cardiac structure. The effect of CDH13 rs4783244 on reduced left ventricular mass index is significantly modified by sleep duration. Short sleep may contribute to the loss of CDH13 locus-mediated cardio-protective effects.
We analyzed the associations of five adiponectin-associated genetic variants with left-ventricular mass index (LVMI), a cardiovascular risk factor, in a population of 559 youth. We found a significant protective association of CDH13 rs4783244 with decreased LVMI, independent of adiponectin and other conventional risk factors. Sleep duration has previously been described as a risk factor for increased LVMI in this population, therefore, we assessed modification of this association in CDH13 by sleep duration, and found that short sleep attenuated the cardio-protective effect of this SNP. Our study provides important insights into pathologic mechanisms and prevention strategies for early risk of cardiovascular disease.
Collapse
Affiliation(s)
- Ge Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Dan Feng
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China
| | - Yonghui Wang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China
| | - Junling Fu
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Lanwen Han
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China
| | - Lujiao Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China
| | - Struan F A Grant
- Division of Endocrinology, The Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania; Philadelphia, PA 19104, USA; Division of Human Genetics, The Children's Hospital of Philadelphia Research Institute, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mingyao Li
- Departments of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ming Li
- Department of Endocrinology, Key Laboratory of Endocrinology, National Health and Family Planning Commission, Peking Union Medical College Hospital, Chinese Academy of Medical Science, Beijing 100730, China.
| | - Shan Gao
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, China.
| |
Collapse
|
14
|
Russo A, Di Gaetano C, Cugliari G, Matullo G. Advances in the Genetics of Hypertension: The Effect of Rare Variants. Int J Mol Sci 2018; 19:E688. [PMID: 29495593 PMCID: PMC5877549 DOI: 10.3390/ijms19030688] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 02/19/2018] [Accepted: 02/26/2018] [Indexed: 12/22/2022] Open
Abstract
Worldwide, hypertension still represents a serious health burden with nine million people dying as a consequence of hypertension-related complications. Essential hypertension is a complex trait supported by multifactorial genetic inheritance together with environmental factors. The heritability of blood pressure (BP) is estimated to be 30-50%. A great effort was made to find genetic variants affecting BP levels through Genome-Wide Association Studies (GWAS). This approach relies on the "common disease-common variant" hypothesis and led to the identification of multiple genetic variants which explain, in aggregate, only 2-3% of the genetic variance of hypertension. Part of the missing genetic information could be caused by variants too rare to be detected by GWAS. The use of exome chips and Next-Generation Sequencing facilitated the discovery of causative variants. Here, we report the advances in the detection of novel rare variants, genes, and/or pathways through the most promising approaches, and the recent statistical tests that have emerged to handle rare variants. We also discuss the need to further support rare novel variants with replication studies within larger consortia and with deeper functional studies to better understand how new genes might improve patient care and the stratification of the response to antihypertensive treatments.
Collapse
Affiliation(s)
- Alessia Russo
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy.
- Italian Institute for Genomic Medicine (IIGM, Formerly HuGeF), 10126 Turin, Italy.
| | - Cornelia Di Gaetano
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy.
- Italian Institute for Genomic Medicine (IIGM, Formerly HuGeF), 10126 Turin, Italy.
| | - Giovanni Cugliari
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy.
- Italian Institute for Genomic Medicine (IIGM, Formerly HuGeF), 10126 Turin, Italy.
| | - Giuseppe Matullo
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy.
- Italian Institute for Genomic Medicine (IIGM, Formerly HuGeF), 10126 Turin, Italy.
| |
Collapse
|
15
|
Li Y, Li C, Ma Q, Zhang Y, Yao Y, Liu S, Zhang X, Hong C, Tan F, Shi L, Yao Y. Genetic variation in CDH13 gene was associated with non-small cell lung cancer (NSCLC): A population-based case-control study. Oncotarget 2017; 9:881-891. [PMID: 29416663 PMCID: PMC5787520 DOI: 10.18632/oncotarget.22971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/15/2017] [Indexed: 12/27/2022] Open
Abstract
Cadherin 13 (CDH13, T-cadherin, H-cadherin) has been identified as an anti-oncogene in various cancers. Recent studies have reported that downregulation of H-cadherin in cancers is associated with CDH13 promoter hypermethylation, which could be affected by the single nucleotide polymorphisms (SNPs) near CpG sites in the CDH13 promoter. In the current study, we investigated and analyzed the association of seven SNPs (rs11646213, rs12596316, rs3865188, rs12444338, rs4783244, rs12051272 and rs7195409) with non-small cell lung cancer (NSCLC) using logistic regression analysis. SNPs rs11646213, rs12596316, rs3865188 and rs12444338 are located in the promoter region, rs4783244 and rs12051272 are located in intron 1, and rs7195409 is located in intron 7. A total of 454 patients with NSCLC were placed into a NSCLC group and 444 healthy controls were placed into a control group, all participants were recruited to genotype the SNPs using Taqman assay. Our results showed that the allelic frequencies of rs11646213 were significantly different between NSCLC and control groups (P = 0.006). In addition, the association analysis of these SNPs stratified into NSCLC pathologic stages I+II and III+IV showed that the allelic frequencies rs7195409 had a significant difference between NSCLC pathologic stages I+II and III+IV (P = 0.006). Our results indicated that the rs11646213 and rs7195409 in CDH13 could be associated with NSCLC or its pathologic stages in the Chinese Han population.
Collapse
Affiliation(s)
- Yingfu Li
- Department of Geriatrics, The No.1 Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Chuanyin Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Qianli Ma
- Department of Thoracic Surgery, The No.3 Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Yu Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yueting Yao
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Shuyuan Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Xinwen Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Chao Hong
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Fang Tan
- Department of Geriatrics, The No.1 Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Li Shi
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| | - Yufeng Yao
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, 650118, China
| |
Collapse
|
16
|
IRS1 DNA promoter methylation and expression in human adipose tissue are related to fat distribution and metabolic traits. Sci Rep 2017; 7:12369. [PMID: 28959056 PMCID: PMC5620072 DOI: 10.1038/s41598-017-12393-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 09/07/2017] [Indexed: 01/08/2023] Open
Abstract
The SNP variant rs2943650 near IRS1 gene locus was previously associated with decreased body fat and IRS1 gene expression as well as an adverse metabolic profile in humans. Here, we hypothesize that these effects may be mediated by an interplay with epigenetic alterations. We measured IRS1 promoter DNA methylation and mRNA expression in paired human subcutaneous and omental visceral adipose tissue samples (SAT and OVAT) from 146 and 41 individuals, respectively. Genotyping of rs2943650 was performed in all individuals (N = 146). We observed a significantly higher IRS1 promoter DNA methylation in OVAT compared to SAT (N = 146, P = 8.0 × 10−6), while expression levels show the opposite effect direction (N = 41, P = 0.011). OVAT and SAT methylation correlated negatively with IRS1 gene expression in obese subjects (N = 16, P = 0.007 and P = 0.010). The major T-allele is related to increased DNA methylation in OVAT (N = 146, P = 0.019). Finally, DNA methylation and gene expression in OVAT correlated with anthropometric traits (waist- circumference waist-to-hip ratio) and parameters of glucose metabolism in obese individuals. Our data suggest that the association between rs2943650 near the IRS1 gene locus with clinically relevant variables may at least be modulated by changes in DNA methylation that translates into altered IRS1 gene expression.
Collapse
|
17
|
Samy MD, Yavorski JM, Mauro JA, Blanck G. Impact of SNPs on CpG Islands in the MYC and HRAS oncogenes and in a wide variety of tumor suppressor genes: A multi-cancer approach. Cell Cycle 2017; 15:1572-8. [PMID: 27074591 DOI: 10.1080/15384101.2016.1164360] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Single nucleotide polymorphisms (SNPs) that occur within CpG Islands may lead to increased hypermethylation if a SNP allele has the potential to form a CpG dinucleotide, as well as potentially lead to hypomethylation if a SNP allele eliminates a CpG dinucleotide. We analyzed CpG-related SNP allele frequencies in whole genome sequences (WGS) across 5 TCGA cancer datasets, thereby exploiting a more recent appreciation for signaling pathway degeneracy in cancer. The cancer data sets were analyzed for SNPs in CpG islands associated with the oncogenes, HRAS and MYC, and in the CpG islands associated with the tumor suppressor genes, APC, DCC, and RB1. We determined that one SNP allele (rs3824120) in a CpG island associated with MYC which eliminated a CpG was more common in the cancer datasets than in the 100Genomes databases (p < 0.01). For HRAS, 2 SNP alleles (rs112690925, rs7939028) that created CpG's occurred significantly less frequently in the cancer data sets than in the general SNP databases (e.g., rs7939028, p < 0.0002, in comparison with AllSNPs(142)). Also, one SNP allele (rs4940177) that created a CpG in a CpG island associated with the DCC tumor suppressor gene, was more common in the cancer datasets (p < 0.0007). To understand a broader picture of the potential of SNP alleles to create CpG's in CpG islands of tumor suppressor genes, we developed a scripted algorithm to assess the SNP alleles associated with the CpG islands of 43 tumor suppressor genes. The following tumor suppressor genes have the possibility of significant, percent increases in their CpG counts, depending on which SNP allele(s) is present: VHL, BRCA1, BRCA2, CHEK2, PTEN and RB1.
Collapse
Affiliation(s)
- Mohammad D Samy
- a Department of Molecular Medicine , Morsani College of Medicine, University of South Florida , Tampa , FL , USA
| | - John M Yavorski
- a Department of Molecular Medicine , Morsani College of Medicine, University of South Florida , Tampa , FL , USA
| | - James A Mauro
- a Department of Molecular Medicine , Morsani College of Medicine, University of South Florida , Tampa , FL , USA
| | - George Blanck
- a Department of Molecular Medicine , Morsani College of Medicine, University of South Florida , Tampa , FL , USA.,b Immunology Program, Moffitt Cancer Center and Research Institute , Tampa , FL , USA
| |
Collapse
|
18
|
Aslibekyan S, Do AN, Xu H, Li S, Irvin MR, Zhi D, Tiwari HK, Absher DM, Shuldiner AR, Zhang T, Chen W, Tanner K, Hong C, Mitchell BD, Berenson G, Arnett DK. CPT1A methylation is associated with plasma adiponectin. Nutr Metab Cardiovasc Dis 2017; 27:225-233. [PMID: 28139377 PMCID: PMC5330786 DOI: 10.1016/j.numecd.2016.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 10/24/2016] [Accepted: 11/14/2016] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIMS Adiponectin, an adipose-secreted protein that has been linked to insulin sensitivity, plasma lipids, and inflammatory patterns, is an established biomarker for metabolic health. Despite clinical relevance and high heritability, the determinants of plasma adiponectin levels remain poorly understood. METHODS AND RESULTS We conducted the first epigenome-wide cross-sectional study of adiponectin levels using methylation data on 368,051 cytosine-phosphate-guanine (CpG) sites in CD4+ T-cells from the Genetics of Lipid Lowering Drugs and Diet Network (GOLDN, n = 991). We fit linear mixed models, adjusting for age, sex, study site, T-cell purity, and family. We have identified a positive association (regression coefficient ± SE = 0.01 ± 0.001, P = 3.4 × 10-13) between plasma adiponectin levels and methylation of a CpG site in CPT1A, a key player in fatty acid metabolism. The association was replicated (n = 474, P = 0.0009) in whole blood samples from the Amish participants of the Heredity and Phenotype Intervention (HAPI) Heart Study as well as White (n = 592, P = 0.0005) but not Black (n = 243, P = 0.18) participants of the Bogalusa Heart Study (BHS). The association remained significant upon adjusting for BMI and smoking in GOLDN and HAPI but not BHS. We also identified associations between methylation loci in RNF145 and UFM1 and plasma adiponectin in GOLDN and White BHS participants, although the association was not robust to adjustment for BMI or smoking. CONCLUSION We have identified and replicated associations between several biologically plausible loci and plasma adiponectin. These findings support the importance of epigenetic processes in metabolic traits, laying the groundwork for future translational applications.
Collapse
Affiliation(s)
- S Aslibekyan
- Department of Epidemiology, University of Alabama at Birmingham, USA.
| | - A N Do
- Department of Epidemiology, University of Alabama at Birmingham, USA
| | - H Xu
- Department of Medicine, University of Maryland School of Medicine, USA
| | - S Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, USA
| | - M R Irvin
- Department of Epidemiology, University of Alabama at Birmingham, USA
| | - D Zhi
- Department of Biostatistics, University of Alabama at Birmingham, USA
| | - H K Tiwari
- Department of Biostatistics, University of Alabama at Birmingham, USA
| | - D M Absher
- HudsonAlpha Institute for Biotechnology, USA
| | - A R Shuldiner
- Department of Medicine, University of Maryland School of Medicine, USA
| | - T Zhang
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, USA
| | - W Chen
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, USA
| | - K Tanner
- Department of Medicine, University of Maryland School of Medicine, USA
| | - C Hong
- Department of Medicine, University of Maryland School of Medicine, USA
| | - B D Mitchell
- Department of Medicine, University of Maryland School of Medicine, USA; Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - G Berenson
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, USA
| | - D K Arnett
- Department of Epidemiology, University of Alabama at Birmingham, USA
| |
Collapse
|
19
|
Nicolas A, Aubert R, Bellili-Muñoz N, Balkau B, Bonnet F, Tichet J, Velho G, Marre M, Roussel R, Fumeron F. T-cadherin gene variants are associated with type 2 diabetes and the Fatty Liver Index in the French population. DIABETES & METABOLISM 2017; 43:33-39. [DOI: 10.1016/j.diabet.2016.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/26/2016] [Accepted: 05/10/2016] [Indexed: 11/30/2022]
|
20
|
Natsukawa T, Maeda N, Fukuda S, Yamaoka M, Fujishima Y, Nagao H, Sato F, Nishizawa H, Sawano H, Hayashi Y, Funahashi T, Kai T, Shimomura I. Significant Association of Serum Adiponectin and Creatine Kinase-MB Levels in ST-Segment Elevation Myocardial Infarction. J Atheroscler Thromb 2017; 24:793-803. [PMID: 28100880 PMCID: PMC5556187 DOI: 10.5551/jat.38232] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Aims: Adiponectin, an adipocyte-specific secretory protein, abundantly exists in the blood stream while its concentration paradoxically decreases in obesity. Hypoadiponectinemia is one of risks of cardiovascular diseases. However, impact of serum adiponectin concentration on acute ischemic myocardial damages has not been fully clarified. The present study investigated the association of serum adiponectin and creatine kinase (CK)-MB levels in subjects with ST-segment elevation myocardial infarction (STEMI). Methods: This study is a physician-initiated observational study and is also registered with the University Hospital Medical Information Network (Number: UMIN 000014418). Patients were admitted to Senri Critical Care Medical Center, given a diagnosis of STEMI, and treated by primary percutaneous coronary intervention (PCI). Finally, 49 patients were enrolled and the association of serum adiponectin, CK-MB, and clinical features were mainly analyzed. Results: Serum adiponectin levels decreased rapidly and reached the bottom at 24 hours after recanalization. Such reduction of serum adiponectin was inversely correlated with the area under the curve (AUC) of serum CK-MB (p = 0.013). Serum adiponectin concentrations were inversely correlated with AUC of serum CK-MB. In multivariate analysis, serum adiponectin concentration on admission (p = 0.002) and collateral (p = 0.037) were significantly and independently correlated with serum AUC of CK-MB. Conclusion: Serum AUC of CK-MB in STEMI subjects was significantly associated with serum adiponectin concentration on admission and reduction of serum adiponectin levels from baseline to bottom. The present study may provide a possibility that serum adiponectin levels at acute phase are useful in the prediction for prognosis after PCI-treated STEMI subjects.
Collapse
Affiliation(s)
- Tomoaki Natsukawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University.,Senri Critical Care Medical Center, Osaka Saiseikai Senri Hospital
| | - Norikazu Maeda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University
| | - Shiro Fukuda
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University
| | - Masaya Yamaoka
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University
| | - Yuya Fujishima
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University
| | - Hirofumi Nagao
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University
| | - Fumi Sato
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University
| | - Hitoshi Nishizawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University
| | - Hirotaka Sawano
- Senri Critical Care Medical Center, Osaka Saiseikai Senri Hospital
| | - Yasuyuki Hayashi
- Senri Critical Care Medical Center, Osaka Saiseikai Senri Hospital
| | - Tohru Funahashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University
| | - Tatsuro Kai
- Senri Critical Care Medical Center, Osaka Saiseikai Senri Hospital
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University
| |
Collapse
|
21
|
Abstract
Coronary artery disease (or coronary heart disease), is the leading cause of mortality in many of the developing as well as the developed countries of the world. Cholesterol-enriched plaques in the heart's blood vessels combined with inflammation lead to the lesion expansion, narrowing of blood vessels, reduced blood flow, and may subsequently cause lesion rupture and a heart attack. Even though several environmental risk factors have been established, such as high LDL-cholesterol, diabetes, and high blood pressure, the underlying genetic composition may substantially modify the disease risk; hence, genome composition and gene-environment interactions may be critical for disease progression. Ongoing scientific efforts have seen substantial advancements related to the fields of genetics and genomics, with the major breakthroughs yet to come. As genomics is the most rapidly advancing field in the life sciences, it is important to present a comprehensive overview of current efforts. Here, we present a summary of various genetic and genomics assays and approaches applied to coronary artery disease research.
Collapse
Affiliation(s)
- Milos Pjanic
- Department of Medicine, Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305-5233, USA
| | - Clint L Miller
- Department of Medicine, Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305-5233, USA
| | - Robert Wirka
- Department of Medicine, Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305-5233, USA
| | - Juyong B Kim
- Department of Medicine, Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305-5233, USA
| | - Daniel M DiRenzo
- Department of Medicine, Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305-5233, USA
| | - Thomas Quertermous
- Department of Medicine, Division of Cardiovascular Medicine, Cardiovascular Institute, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA, 94305-5233, USA.
| |
Collapse
|
22
|
Xu Y, Li X, Wang H, Xie P, Yan X, Bai Y, Zhang T. Hypermethylation of CDH13, DKK3 and FOXL2 promoters and the expression of EZH2 in ovary granulosa cell tumors. Mol Med Rep 2016; 14:2739-45. [DOI: 10.3892/mmr.2016.5521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 05/16/2016] [Indexed: 11/05/2022] Open
|
23
|
Balatskaya MN, Balatskii AV, Sharonov GV, Tkachuk VA. T-cadherin as a novel receptor regulating metabolism in the blood vessel and heart cells: from structure to function. J EVOL BIOCHEM PHYS+ 2016. [DOI: 10.1134/s0022093016020010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
24
|
Lemche E, Chaban OS, Lemche AV. Neuroendocrinological and Epigenetic Mechanisms Subserving Autonomic Imbalance and HPA Dysfunction in the Metabolic Syndrome. Front Neurosci 2016; 10:142. [PMID: 27147943 PMCID: PMC4830841 DOI: 10.3389/fnins.2016.00142] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/21/2016] [Indexed: 12/18/2022] Open
Abstract
Impact of environmental stress upon pathophysiology of the metabolic syndrome (MetS) has been substantiated by epidemiological, psychophysiological, and endocrinological studies. This review discusses recent advances in the understanding of causative roles of nutritional factors, sympathomedullo-adrenal (SMA) and hypothalamic-pituitary adrenocortical (HPA) axes, and adipose tissue chronic low-grade inflammation processes in MetS. Disturbances in the neuroendocrine systems for leptin, melanocortin, and neuropeptide Y (NPY)/agouti-related protein systems have been found resulting directly in MetS-like conditions. The review identifies candidate risk genes from factors shown critical for the functioning of each of these neuroendocrine signaling cascades. In its meta-analytic part, recent studies in epigenetic modification (histone methylation, acetylation, phosphorylation, ubiquitination) and posttranscriptional gene regulation by microRNAs are evaluated. Several studies suggest modification mechanisms of early life stress (ELS) and diet-induced obesity (DIO) programming in the hypothalamic regions with populations of POMC-expressing neurons. Epigenetic modifications were found in cortisol (here HSD11B1 expression), melanocortin, leptin, NPY, and adiponectin genes. With respect to adiposity genes, epigenetic modifications were documented for fat mass gene cluster APOA1/C3/A4/A5, and the lipolysis gene LIPE. With regard to inflammatory, immune and subcellular metabolism, PPARG, NKBF1, TNFA, TCF7C2, and those genes expressing cytochrome P450 family enzymes involved in steroidogenesis and in hepatic lipoproteins were documented for epigenetic modifications.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London London, UK
| | - Oleg S Chaban
- Section of Psychosomatic Medicine, Bogomolets National Medical University Kiev, Ukraine
| | - Alexandra V Lemche
- Department of Medical Science, Institute of Clinical Research Berlin, Germany
| |
Collapse
|
25
|
Schleinitz D. Genetic Determination of Serum Levels of Diabetes-Associated Adipokines. Rev Diabet Stud 2016; 12:277-98. [PMID: 26859657 PMCID: PMC5275755 DOI: 10.1900/rds.2015.12.277] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Adipose tissue secretes an abundance of proteins. Some of these proteins are known as adipokines and adipose-derived hormones which have been linked with metabolic disorders, including type 2 diabetes, and even with cancer. Variance in serum adipokine concentration is often closely associated with an increase (obesity) or decrease (lipodystrophy) in fat tissue mass, and it is affected by age, gender, and localization of the adipose tissue. However, there may be genetic variants which, in consequence, influence the serum concentration of a certain adipokine, and thereby promote metabolic disturbances or, with regard to the "protective" allele, exert beneficial effects. This review focuses on the genetic determination of serum levels of the following adipokines: adiponectin, chemerin, leptin, progranulin, resistin, retinol binding protein 4, vaspin, adipsin, apelin, and omentin. The article reports on the latest findings from genome-wide association studies (GWAS) and candidate gene studies, showing variants located in/nearby the adipokine genes and other (non-receptor) genes. An extra chapter highlights adipokine-receptor variants. Epigenetic studies on adipokines are also addressed.
Collapse
Affiliation(s)
- Dorit Schleinitz
- Integrated Research and Treatment Center AdiposityDiseases, University of Leipzig, Liebigstr. 21, 04103 Leipzig, Germany
| |
Collapse
|
26
|
Ladd-Acosta C, Fallin MD. The role of epigenetics in genetic and environmental epidemiology. Epigenomics 2015; 8:271-83. [PMID: 26505319 DOI: 10.2217/epi.15.102] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Epidemiology is the branch of science that investigates the causes and distribution of disease in populations in order to provide preventative measures and promote human health. The fields of genetic and environmental epidemiology primarily seek to identify genetic and environmental risk factors for disease, respectively. Epigenetics is emerging as an important piece of molecular data to include in these studies because it can provide mechanistic insights into genetic and environmental risk factors for disease, identify potential intervention targets, provide biomarkers of exposure, illuminate gene-environment interactions and help localize disease-relevant genomic regions. Here, we describe the importance of including epigenetics in genetic and environmental epidemiology studies, provide a conceptual framework when considering epigenetic data in population-based studies and touch upon the many challenges that lie ahead.
Collapse
Affiliation(s)
- Christine Ladd-Acosta
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, MD 21205, USA
| | - M Daniele Fallin
- Department of Mental Health, Johns Hopkins School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|