251
|
Feil R, Lehners M, Stehle D, Feil S. Visualising and understanding cGMP signals in the cardiovascular system. Br J Pharmacol 2021; 179:2394-2412. [PMID: 33880767 DOI: 10.1111/bph.15500] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/14/2021] [Accepted: 04/01/2021] [Indexed: 12/21/2022] Open
Abstract
cGMP is an important signalling molecule in humans. Fluorescent cGMP biosensors have emerged as powerful tools for the sensitive analysis of cGMP pathways at the single-cell level. Here, we briefly outline cGMP's multifaceted role in (patho)physiology and pharmacotherapy. Then we summarise what new insights cGMP imaging has provided into endogenous cGMP signalling and drug action, with a focus on the cardiovascular system. Indeed, the use of cGMP biosensors has led to several conceptual advances, such as the discovery of local, intercellular and mechanosensitive cGMP signals. Importantly, single-cell imaging can provide valuable information about the heterogeneity of cGMP signals within and between individual cells of an isolated cell population or tissue. We also discuss current challenges and future directions of cGMP imaging, such as the direct visualisation of cGMP microdomains, simultaneous monitoring of cGMP and other signalling molecules and, ultimately, cGMP imaging in tissues and animals under close-to-native conditions.
Collapse
Affiliation(s)
- Robert Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Moritz Lehners
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Daniel Stehle
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| | - Susanne Feil
- Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany
| |
Collapse
|
252
|
Xie M, Yuan S, Zeng Y, Zheng C, Yang Y, Dong Y, He Q. ATP2B1 gene polymorphisms rs2681472 and rs17249754 are associated with susceptibility to hypertension and blood pressure levels: A systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e25530. [PMID: 33847678 PMCID: PMC8052043 DOI: 10.1097/md.0000000000025530] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/23/2021] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE The present study aimed to conduct a systematic review and meta-analysis to evaluate the relationships between ATP2B1 gene polymorphisms with blood pressure (BP) level and susceptibility to hypertension. METHODS PubMed, Web of Science, Embase and China National Knowledge Infrastructure (CNKI) Databases were systematically searched by 2 independent researchers to screen studies on ATP2B1 gene polymorphisms and BP related phenotypes. The records retrieval period was limited from the formation of the database to March 4, 2021. Pooled odds rations (ORs) or β and their 95% confidence intervals (95%CI) were calculated to assess the association between ATP2B1 gene polymorphisms and the risk of hypertension or BP levels. Publication bias and sensitivity analysis were conducted to find potential bias. All the statistical analysis were conducted with Stata version 11.0 software. RESULTS A total of 15 articles were ultimately included in the present study, including 15 polymorphisms of ATP2B1 gene. Nine articles (N = 65,362) reported the polymorphism rs17249754, and 7 articles(N = 91,997) reported rs2681472 (both loci were reported in 1 article). Meta-analysis showed that rs17249754 (G/A) and rs2681472 (A/G) were associated with the susceptibility to hypertension (rs17249754: OR = 1.19, 95%CI: 1.10-1.28; rs2681472: OR = 1.15, 95%CI: 1.12-1.17), and were positively associated with systolic BP (SBP) and diastolic blood pressure (DBP) (rs17249754: SBP, β=1.01, 95%CI: 0.86-1.16, DBP, β=0.48, 95%CI: 0.30-0.66; rs2681472: SBP, β=0.92, 95%CI: 0.77-1.07, DBP, β=0.50, 95%CI: 0.42-0.58) in the additive genetic model. Subgroup analysis stratified by race, population, sample size, and BP measurement method revealed that the association between A allele in rs2681472 polymorphism and risk of hypertension was slightly stronger in European (EUR) populations (OR = 1.16, 95%CI: 1.13-1.20) than in East Asians (OR = 1.14, 95%CI: 1.10-1.17). While in East Asians, relation between rs17249754 with risk of hypertension (OR = 1.19, 95%CI: 1.10-1.28) is stronger than rs2681472 (OR = 1.14, 95%CI: 1.10-1.17). CONCLUSIONS Our study demonstrated that ATP2B1 gene polymorphism rs2681472 and rs17249754 were associated with BP levels and the susceptibility to hypertension.
Collapse
Affiliation(s)
- Ming Xie
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha
| | - Shuqian Yuan
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha
| | - Yuan Zeng
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha
| | - Chanjuan Zheng
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha
| | - Yide Yang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Yanhui Dong
- Institute of Child and Adolescent Health, School of Public Health, Peking University Health Science Center, Beijing, China
| | - Quanyuan He
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha
| |
Collapse
|
253
|
Nierenberg JL, Anderson AH, He J, Parsa A, Srivastava A, Cohen JB, Saraf SL, Rahman M, Rosas SE, Kelly TN, CRIC Study Investigators. Association of Blood Pressure Genetic Risk Score with Cardiovascular Disease and CKD Progression: Findings from the CRIC Study. KIDNEY360 2021; 2:1251-1260. [PMID: 35369652 PMCID: PMC8676389 DOI: 10.34067/kid.0007632020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/30/2021] [Indexed: 02/04/2023]
Abstract
Background In the general population, genetic risk for high BP has been associated with cardiovascular disease, but not kidney function or incident CKD. These relationships have not been studied longitudinally in participants with CKD. We examined whether BP genetic risk predicts cardiovascular disease and kidney disease progression in patients with CKD. Methods We included 1493 African- and 1581 European-ancestry participants from the Chronic Renal Insufficiency Cohort who were followed for 12 years. We examined associations of BP genetic risk scores with development of cardiovascular disease (myocardial infarction, congestive heart failure, or stroke) and CKD progression (incident ESKD or halving of eGFR) using Cox proportional hazards models. Analyses were stratified by race and included adjustment for age, sex, study site, and ancestry principal components. Results Among European-ancestry participants, each SD increase in systolic BP and pulse pressure genetic risk score conferred a 15% (95% CI, 4% to 27%) and 11% (95% CI, 1% to 23%), respectively, higher risk of cardiovascular disease, with a similar, marginally significant trend for diastolic BP. Among African-ancestry participants, each SD increase in systolic and diastolic BP genetic risk score conferred a 10% (95% CI, 1% to 20%) and 9% (95% CI, 0% to 18%), respectively, higher risk of cardiovascular disease. Higher genetic risk was not associated with CKD progression. Conclusions Genetic risk for elevation in BP was associated with increased risk of cardiovascular disease, but not CKD progression.
Collapse
Affiliation(s)
- Jovia L. Nierenberg
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Amanda H. Anderson
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana,Department of Medicine, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | - Afshin Parsa
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, The National Institutes of Health, Bethesda, Maryland
| | - Anand Srivastava
- Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jordana B. Cohen
- Renal-Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Santosh L. Saraf
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Sickle Cell Center, University of Illinois at Chicago, Chicago, Illinois
| | - Mahboob Rahman
- Division of Nephrology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Sylvia E. Rosas
- Kidney and Hypertension Unit, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Tanika N. Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana
| | | |
Collapse
|
254
|
Mabhida SE, Mashatola L, Kaur M, Sharma JR, Apalata T, Muhamed B, Benjeddou M, Johnson R. Hypertension in African Populations: Review and Computational Insights. Genes (Basel) 2021; 12:genes12040532. [PMID: 33917487 PMCID: PMC8067483 DOI: 10.3390/genes12040532] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 01/11/2023] Open
Abstract
Hypertension (HTN) is a persistent public health problem affecting approximately 1.3 billion individuals globally. Treatment-resistant hypertension (TRH) is defined as high blood pressure (BP) in a hypertensive patient that remains above goal despite use of ≥3 antihypertensive agents of different classes including a diuretic. Despite a plethora of treatment options available, only 31.0% of individuals have their HTN controlled. Interindividual genetic variability to drug response might explain this disappointing outcome because of genetic polymorphisms. Additionally, the poor knowledge of pathophysiological mechanisms underlying hypertensive disease and the long-term interaction of antihypertensive drugs with blood pressure control mechanisms further aggravates the problem. Furthermore, in Africa, there is a paucity of pharmacogenomic data on the treatment of resistant hypertension. Therefore, identification of genetic signals having the potential to predict the response of a drug for a given individual in an African population has been the subject of intensive investigation. In this review, we aim to systematically extract and discuss African evidence on the genetic variation, and pharmacogenomics towards the treatment of HTN. Furthermore, in silico methods are utilized to elucidate biological processes that will aid in identifying novel drug targets for the treatment of resistant hypertension in an African population. To provide an expanded view of genetic variants associated with the development of HTN, this study was performed using publicly available databases such as PubMed, Scopus, Web of Science, African Journal Online, PharmGKB searching for relevant papers between 1984 and 2020. A total of 2784 articles were reviewed, and only 42 studies were included following the inclusion criteria. Twenty studies reported associations with HTN and genes such as AGT (rs699), ACE (rs1799752), NOS3 (rs1799983), MTHFR (rs1801133), AGTR1 (rs5186), while twenty-two studies did not show any association within the African population. Thereafter, an in silico predictive approach was utilized to identify several genes including CLCNKB, CYPB11B2, SH2B2, STK9, and TBX5 which may act as potential drug targets because they are involved in pathways known to influence blood pressure. Next, co-expressed genes were identified as they are controlled by the same transcriptional regulatory program and may potentially be more effective as multiple drug targets in the treatment regimens for HTN. Genes belonging to the co-expressed gene cluster, ACE, AGT, AGTR1, AGTR2, and NOS3 as well as CSK and ADRG1 showed enrichment of G-protein-coupled receptor activity, the classical targets of drug discovery, which mediate cellular signaling processes. The latter is of importance, as the targeting of co-regulatory gene clusters will allow for the development of more effective HTN drug targets that could decrease the prevalence of both controlled and TRH.
Collapse
Affiliation(s)
- Sihle E. Mabhida
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (S.E.M.); (J.R.S.)
- Department of Biotechnology, Faculty of Natural Science, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa;
| | - Lebohang Mashatola
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Johannesburg 2050, South Africa; (L.M.); (M.K.)
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Johannesburg 2050, South Africa; (L.M.); (M.K.)
| | - Jyoti R. Sharma
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (S.E.M.); (J.R.S.)
| | - Teke Apalata
- Division of Medical Microbiology, Department of Laboratory-Medicine and Pathology, Faculty of Health Sciences, Walter Sisulu University and National Health Laboratory Services, Mthatha 5100, South Africa;
| | - Babu Muhamed
- Hatter Institute for Cardiovascular Diseases Research in Africa, Department of Medicine, University of Cape Town, Cape Town 7535, South Africa;
- Children’s National Health System, Division of Cardiology, Washington, DC 20010, USA
| | - Mongi Benjeddou
- Department of Biotechnology, Faculty of Natural Science, University of the Western Cape, Private Bag X17, Bellville, Cape Town 7535, South Africa;
| | - Rabia Johnson
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; (S.E.M.); (J.R.S.)
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
- Correspondence: ; Tel.: +27-21-938-0866
| |
Collapse
|
255
|
Madhur MS, Elijovich F, Alexander MR, Pitzer A, Ishimwe J, Van Beusecum JP, Patrick DM, Smart CD, Kleyman TR, Kingery J, Peck RN, Laffer CL, Kirabo A. Hypertension: Do Inflammation and Immunity Hold the Key to Solving this Epidemic? Circ Res 2021; 128:908-933. [PMID: 33793336 PMCID: PMC8023750 DOI: 10.1161/circresaha.121.318052] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Elevated cardiovascular risk including stroke, heart failure, and heart attack is present even after normalization of blood pressure in patients with hypertension. Underlying immune cell activation is a likely culprit. Although immune cells are important for protection against invading pathogens, their chronic overactivation may lead to tissue damage and high blood pressure. Triggers that may initiate immune activation include viral infections, autoimmunity, and lifestyle factors such as excess dietary salt. These conditions activate the immune system either directly or through their impact on the gut microbiome, which ultimately produces chronic inflammation and hypertension. T cells are central to the immune responses contributing to hypertension. They are activated in part by binding specific antigens that are presented in major histocompatibility complex molecules on professional antigen-presenting cells, and they generate repertoires of rearranged T-cell receptors. Activated T cells infiltrate tissues and produce cytokines including interleukin 17A, which promote renal and vascular dysfunction and end-organ damage leading to hypertension. In this comprehensive review, we highlight environmental, genetic, and microbial associated mechanisms contributing to both innate and adaptive immune cell activation leading to hypertension. Targeting the underlying chronic immune cell activation in hypertension has the potential to mitigate the excess cardiovascular risk associated with this common and deadly disease.
Collapse
Affiliation(s)
- Meena S. Madhur
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center
- Department of Molecular Physiology and Biophysics, Vanderbilt University
| | - Fernando Elijovich
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matthew R. Alexander
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center
| | - Ashley Pitzer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jeanne Ishimwe
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Justin P. Van Beusecum
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - David M. Patrick
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center
| | - Charles D. Smart
- Department of Molecular Physiology and Biophysics, Vanderbilt University
| | - Thomas R. Kleyman
- Departments of Medicine, Cell Biology, Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Justin Kingery
- Center for Global Health, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Weill Bugando School of Medicine, Mwanza, Tanzania
| | - Robert N. Peck
- Center for Global Health, Weill Cornell Medical College, New York, NY, USA
- Department of Medicine, Weill Bugando School of Medicine, Mwanza, Tanzania
- Mwanza Intervention Trials Unit (MITU), Mwanza, Tanzania
| | - Cheryl L. Laffer
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University
| |
Collapse
|
256
|
Abstract
The known genetic architecture of blood pressure now comprises >30 genes, with rare variants resulting in monogenic forms of hypertension or hypotension and >1,477 common single-nucleotide polymorphisms (SNPs) being associated with the blood pressure phenotype. Monogenic blood pressure syndromes predominantly involve the renin-angiotensin-aldosterone system and the adrenal glucocorticoid pathway, with a smaller fraction caused by neuroendocrine tumours of the sympathetic and parasympathetic nervous systems. The SNPs identified in genome-wide association studies (GWAS) as being associated with the blood pressure phenotype explain only approximately 27% of the 30-50% estimated heritability of blood pressure, and the effect of each SNP on the blood pressure phenotype is small. A paucity of SNPs from GWAS are mapped to known genes causing monogenic blood pressure syndromes. For example, a GWAS signal mapped to the gene encoding uromodulin has been shown to affect blood pressure by influencing sodium homeostasis, and the effects of another GWAS signal were mediated by endothelin. However, the majority of blood pressure-associated SNPs show pleiotropic associations. Unravelling these associations can potentially help us to understand the underlying biological pathways. In this Review, we appraise the current knowledge of blood pressure genomics, explore the causal pathways for hypertension identified in Mendelian randomization studies and highlight the opportunities for drug repurposing and pharmacogenomics for the treatment of hypertension.
Collapse
Affiliation(s)
- Sandosh Padmanabhan
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Anna F Dominiczak
- BHF Glasgow Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
257
|
Sharina I, Lezgyieva K, Krutsenko Y, Martin E. Higher susceptibility to heme oxidation and lower protein stability of the rare α 1C517Yβ 1 sGC variant associated with moyamoya syndrome. Biochem Pharmacol 2021; 186:114459. [PMID: 33571505 PMCID: PMC8052303 DOI: 10.1016/j.bcp.2021.114459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/22/2020] [Accepted: 01/29/2021] [Indexed: 12/20/2022]
Abstract
NO sensitive soluble guanylyl cyclase (sGC) plays a key role in mediating physiological functions of NO. Genetic alterations of the GUCY1A3 gene, coding for the α1 subunit of sGC, are associated with several cardiovascular dysfunctions. A rare sGC variant with Cys517 → Tyr substitution in the α1subunit, has been associated with moyamoya disease and achalasia. In this report we characterize the properties of this rare sGC variant. Purified α1C517Yβ1 sGC preserved only ~25% of its cGMP-forming activity and showed an elevated Km for GTP substrate. However, the mutant enzyme retained a high affinity for and robust activation by NO, similar to wild type sGC. Purified α1C517Yβ1 enzyme was more sensitive to specific sGC heme oxidizers and less responsive to heme reducing agents. When expressed in COS7 cells, α1C517Yβ1 sGC showed a much stronger response to cinaciguat or gemfibrozil, which targets apo-sGC or sGC with ferric heme, as compared to its NO response or the relative response of the wild type sGC. A stronger response to cinaciguat was also observed for purified α1C517Yβ1 in the absence of reducing agents. In COS7 cells, αCys517β sGC was less stable than the wild type enzyme under normal conditions and exhibited accelerated degradation upon induction of cellular oxidative stress. We conclude that diminished cGMP-forming activity of this sGC variant is aggravated by its high susceptibility to oxidative stress and diminished protein stability. The combination of these deficiencies contributes to the severity of observed moyamoya and achalasia symptoms in human carriers of this rare α1C517Yβ1 sGC variant.
Collapse
Affiliation(s)
- Iraida Sharina
- University of Texas Health Science Center, McGovern Medical School, Department of Internal Medicine, Division of Cardiology, United States
| | - Karina Lezgyieva
- School of Science and Technology, Nazarbayev University, Astana, Kazakhstan
| | | | - Emil Martin
- University of Texas Health Science Center, McGovern Medical School, Department of Internal Medicine, Division of Cardiology, United States.
| |
Collapse
|
258
|
Sun JY, Zhang H, Zhang Y, Wang L, Sun BL, Gao F, Liu G. Impact of serum calcium levels on total body bone mineral density: A mendelian randomization study in five age strata. Clin Nutr 2021; 40:2726-2733. [PMID: 33933738 DOI: 10.1016/j.clnu.2021.03.012] [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: 07/10/2020] [Revised: 03/01/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Mendelian randomization (MR) studies have reported the causal association between serum calcium levels and bone mineral density (BMD). The results showed that genetically increased serum calcium levels in individuals with normal calcium levels did not increase BMD and could even reduce BMD. However, whether there are differences in the association between serum calcium and BMD in different age strata remains unclear. METHODS We selected eight serum calcium genetic variants with genome-wide significance (P < 5.00E-08) as the potential instrumental variables. We conducted an MR analysis to evaluate the impact of serum calcium levels on total body BMD in five age strata, 0-15, 15-30, 30-45, 45-60, and ≥60 years, using large-scale serum calcium (61,079 individuals) and total body BMD genome-wide association study (66,628 individuals) datasets. For pleiotropy analysis, we used a manual method and four common statistical methods, namely the MR-Egger intercept, MR-PRESSO, heterogeneity, and Steiger filtering tests. For MR analysis, we selected four MR methods, namely inverse-variance weighted, weighted median, MR-Egger, and MR-PRESSO. In addition to the univariable MR analysis, we conducted a multivariate MR analysis taking into account the effect of serum parathyroid hormone levels. RESULTS Univariable MR analysis using the inverse-variance weighted method indicated that per 0.5-mg/dL increase (about 1 standard deviation) in serum calcium levels was statistically significantly associated with reduced total body BMD only in the ≥60 years stratum (effect estimate (beta) = -0.545, 95% confidence interval (CI): -0.892 to -0.198, P = 0.002). The weighted median regression (beta = -0.446, 95% CI: -0.821 to -0.094, P = 1.40E-02) and MR-PRESSO (beta = -0.545, 95% CI: -0.892 to -0.198, P = 0.022) MR methods further supported this suggestive association. The multivariable MR analysis also found a significant association between increased serum calcium levels and reduced total body BMD in the ≥60 years stratum (beta = -0.547, 95% CI: -0.934 to -0.16, P = 0.006). CONCLUSIONS Our results provide genetic evidence that increased serum calcium levels did not improve BMD in the general population and that the elevated serum calcium levels in generally healthy populations, especially in adults older than 60 years, may even reduce the BMD. Our results are comparable with those of recent MR findings.
Collapse
Affiliation(s)
- Jing-Yi Sun
- Shandong Provincial Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250021, China
| | - Haihua Zhang
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China
| | - Yan Zhang
- Department of Pathology, The Affiliated Hospital of Weifang Medical University, Weifang, 261053, China
| | - Longcai Wang
- Department of Anesthesiology, The Affiliated Hospital of Weifang Medical University, Weifang, 261053, China
| | - Bao-Liang Sun
- Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Department of Neurology, Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China
| | - Feng Gao
- Department of Trauma and Emergency Surgeon, The Second Affiliated Hospital, Harbin Medical University, Harbin, China.
| | - Guiyou Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China; Key Laboratory of Cerebral Microcirculation in Universities of Shandong, Department of Neurology, Second Affiliated Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, Shandong, China; National Engineering Laboratory of Internet Medical Diagnosis and Treatment Technology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China; Beijing Key Laboratory of Hypoxia Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
259
|
Zhong Y, Chen L, Li J, Yao Y, Liu Q, Niu K, Ma Y, Xu Y. Integration of summary data from GWAS and eQTL studies identified novel risk genes for coronary artery disease. Medicine (Baltimore) 2021; 100:e24769. [PMID: 33725943 PMCID: PMC7982177 DOI: 10.1097/md.0000000000024769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 01/23/2021] [Indexed: 01/05/2023] Open
Abstract
Several genetic loci have been reported to be significantly associated with coronary artery disease (CAD) by multiple genome-wide association studies (GWAS). Nevertheless, the biological and functional effects of these genetic variants on CAD remain largely equivocal. In the current study, we performed an integrative genomics analysis by integrating large-scale GWAS data (N = 459,534) and 2 independent expression quantitative trait loci (eQTL) datasets (N = 1890) to determine whether CAD-associated risk single nucleotide polymorphisms (SNPs) exert regulatory effects on gene expression. By using Sherlock Bayesian, MAGMA gene-based, multidimensional scaling (MDS), functional enrichment, and in silico permutation analyses for independent technical and biological replications, we highlighted 4 susceptible genes (CHCHD1, TUBG1, LY6G6C, and MRPS17) associated with CAD risk. Based on the protein-protein interaction (PPI) network analysis, these 4 genes were found to interact with each other. We detected a remarkably altered co-expression pattern among these 4 genes between CAD patients and controls. In addition, 3 genes of CHCHD1 (P = .0013), TUBG1 (P = .004), and LY6G6C (P = .038) showed significantly different expressions between CAD patients and controls. Together, we provide evidence to support that these identified genes such as CHCHD1 and TUBG1 are indicative factors of CAD.
Collapse
Affiliation(s)
- Yigang Zhong
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine
| | | | - Jingjing Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou
| | - Yinghao Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou
| | - Qiang Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou
| | - Kaimeng Niu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou
| | - Yunlong Ma
- Institute of Biomedical Big Data, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine
- Zhejiang Chinese Medical University
| |
Collapse
|
260
|
Nipping Adipocyte Inflammation in the Bud. ACTA ACUST UNITED AC 2021; 3. [PMID: 33732506 PMCID: PMC7963359 DOI: 10.20900/immunometab20210012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Adipose tissue inflammation continues to represent a significant area of research in immunometabolism. We have identified a transcription factor, EBF1, which crucially regulates the expression of numerous inflammatory loci in adipocytes. However, EBF1 appears to do so without physically binding to these inflammatory genes. Our research is currently focused on understanding this discrepancy, and we believe that future findings could pave the road for drug development aimed to block adipose inflammation at its source.
Collapse
|
261
|
Li Y, Hong Z. Exposure to the Chinese famine in early life and self-reported arthritis risk in adulthood. PSYCHOL HEALTH MED 2021; 27:1553-1562. [PMID: 33733970 DOI: 10.1080/13548506.2021.1903052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Evidence shows that undernutrition during early life is associated with an increased risk of chronic diseases in adulthood. We aimed to investigate whether exposure to the Chinese famine in the fetal or infant stage was associated with self-reported arthritis risk in adulthood. A total of 3,622 participants were included in the final analysis. Participants were classified into non-, fetal-, and infant-exposed group. Arthritis was self-reported doctor-diagnosed arthritis. A multivariate logistic regression model was used to calculate the odds ratios (ORs) and 95% confidence intervals (CIs) of arthritis. The prevalence of arthritis was 27.07%, 27.74%, and 34.09% among individuals in non-, fetal-, infant-exposed group, respectively. Infant-exposed group (OR: 1.32; 95% CI: 1.12-1.56) had a higher arthritis risk then non-exposed group after adjustment for gender, age, area, education level, smoking status, drinking status, and physical activity. Participants who experienced severe famine during infant and fetal period had higher (41.46%, OR=1.71 and 32.94%, OR=1.36) arthritis risk than those exposed to less severe famine. Exposure to the Chinese famine in early life was associated with an increased risk of arthritis in adulthood, which was partially influenced by some factors (e.g., gender, area, body mass index, and born in severely affected area or not).
Collapse
Affiliation(s)
- Yaru Li
- Department of Nutrition, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhongxin Hong
- Department of Nutrition, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
262
|
Vishnolia KK, Rakovic A, Hoene C, Tarhbalouti K, Aherrahrou Z, Erdmann J. sGC Activity and Regulation of Blood Flow in a Zebrafish Model System. Front Physiol 2021; 12:633171. [PMID: 33716783 PMCID: PMC7946990 DOI: 10.3389/fphys.2021.633171] [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: 11/25/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Soluble guanylyl cyclase (sGC) protein is a heterodimer formed by two subunits encoded by GUCY1A1 and GUCY1B1 genes. The chromosomal locus 4q32.1 harbors both of these genes, which has been previously significantly associated with coronary artery disease, myocardial infarction, and high blood pressure. Blood pressure is influenced by both the environment and genetics and is complemented by several biological pathways. The underlying mechanisms associated with this locus and its genes still need to be investigated. In the current study, we aimed to establish the zebrafish as a model organism to investigate the mechanisms surrounding sGC activity and blood pressure. A zebrafish mutant gucy1a1 line was generated using the CRISPR-Cas9 system by inducing a 4-bp deletion frameshift mutation. This mutation resulted in a reduction of gucy1a1 expression in both heterozygote and homozygote zebrafish. Blood flow parameters (blood flow, arterial pulse, linear velocity, and vessel diameter) investigated in the gucy1a1 mutants showed a significant increase in blood flow and linear velocity, which was augmented in the homozygotes. No significant differences were observed for the blood flow parameters measured from larvae with individual morpholino downregulation of gucy1a1 and gucy1b1, but an increase in blood flow and linear velocity was observed after co-morpholino downregulation of both genes. In addition, the pharmacological sGC stimulator BAY41-2272 rescued the impaired cGMP production in the zebrafish gucy1a1± mutant larvae. Downregulation of cct7 gene did not show any significant difference on the blood flow parameters in both wild-type and gucy1a1± background larvae. In summary, we successfully established a zebrafish platform for investigating sGC-associated pathways and underlying mechanisms in depth. This model system will have further applications, including for potential drug screening experiments.
Collapse
Affiliation(s)
- Krishan K Vishnolia
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany.,University of Lübeck, Lübeck, Germany
| | | | - Celine Hoene
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany.,University of Lübeck, Lübeck, Germany
| | - Karim Tarhbalouti
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany.,University of Lübeck, Lübeck, Germany
| | - Zouhair Aherrahrou
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany.,University of Lübeck, Lübeck, Germany
| | - Jeanette Erdmann
- Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany.,DZHK (German Research Centre for Cardiovascular Research), Partner Site Hamburg/Lübeck/Kiel, Lübeck, Germany.,University of Lübeck, Lübeck, Germany
| |
Collapse
|
263
|
Nazarzadeh M, Pinho-Gomes AC, Bidel Z, Canoy D, Dehghan A, Smith Byrne K, Bennett DA, Smith GD, Rahimi K. Genetic susceptibility, elevated blood pressure, and risk of atrial fibrillation: a Mendelian randomization study. Genome Med 2021; 13:38. [PMID: 33663581 PMCID: PMC7934395 DOI: 10.1186/s13073-021-00849-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 02/09/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Whether elevated blood pressure (BP) is a modifiable risk factor for atrial fibrillation (AF) is not established. We tested (1) whether the association between BP and risk of AF is causal, (2) whether it varies according to individual's genetic susceptibility for AF, and (3) the extent to which specific BP-lowering drugs are expected to reduce this risk. METHODS First, causality of association was assessed through two-sample Mendelian randomization, using data from two independent genome-wide association studies that included a population of one million Europeans in total. Second, the UK Biobank data of 329,237 participants at baseline was used to study the effect of BP on AF according to genetic susceptibility of developing AF. Third, a possible treatment effect with major BP-lowering drug classes on AF risk was predicted through genetic variants in genes encode the therapeutic targets of each drug class. Estimated drug effects were compared with effects on incident coronary heart disease, for which direct trial evidence exists. RESULTS The two-sample Mendelian randomization analysis indicated that, on average, exposure to a higher systolic BP increased the risk of AF by 19% (odds ratio per each 10-mmHg [OR] 1.19 [1.12 to 1.27]). This association was replicated in the UK biobank using individual participant data. However, in a further genetic risk-stratified analysis, there was evidence for a linear gradient in the relative effects of systolic BP on AF; while there was no conclusive evidence of an effect in those with low genetic risk, a strong effect was observed among those with high genetic susceptibility for AF. The comparison of predicted treatment effects using genetic proxies for three main drug classes (angiotensin-converting enzyme inhibitors, beta-blockers, and calcium channel blockers) suggested similar average effects for the prevention of atrial fibrillation and coronary heart disease. CONCLUSIONS The effect of elevated BP on the risk of AF is likely to be causal, suggesting that BP-lowering treatment may be effective in AF prevention. However, average effects masked clinically important variations, with a more pronounced effect in individuals with high genetic susceptibility risk for AF.
Collapse
Affiliation(s)
- Milad Nazarzadeh
- Deep Medicine, Oxford Martin School, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford, OX1 2BQ, UK
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | | | - Zeinab Bidel
- Deep Medicine, Oxford Martin School, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford, OX1 2BQ, UK
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
| | - Dexter Canoy
- Deep Medicine, Oxford Martin School, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford, OX1 2BQ, UK
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Abbas Dehghan
- Department of Biostatistics and Epidemiology, School of Public Health, Imperial College London, London, UK
| | - Karl Smith Byrne
- Deep Medicine, Oxford Martin School, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford, OX1 2BQ, UK
| | - Derrick A Bennett
- Clinical Trial Service Unit and Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | | | - Kazem Rahimi
- Deep Medicine, Oxford Martin School, University of Oxford, 1st Floor, Hayes House, 75 George Street, Oxford, OX1 2BQ, UK.
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
264
|
Shao S, Li XD, Lu YY, Li SJ, Chen XH, Zhou HD, He S, Guo YT, Lu X, Gao PJ, Wang JG. Renal Natriuretic Peptide Receptor-C Deficiency Attenuates NaCl Cotransporter Activity in Angiotensin II-Induced Hypertension. Hypertension 2021; 77:868-881. [PMID: 33486984 DOI: 10.1161/hypertensionaha.120.15636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Genome-wide association studies have identified that NPR-C (natriuretic peptide receptor-C) variants are associated with elevation of blood pressure. However, the mechanism underlying the relationship between NPR-C and blood pressure regulation remains elusive. Here, we investigate whether NPR-C regulates Ang II (angiotensin II)-induced hypertension through sodium transporters activity. Wild-type mice responded to continuous Ang II infusion with an increased renal NPR-C expression. Global NPR-C deficiency attenuated Ang II-induced increased blood pressure both in male and female mice associated with more diuretic and natriuretic responses to a saline challenge. Interestingly, Ang II increased both total and phosphorylation of NCC (NaCl cotransporter) abundance involving in activation of WNK4 (with-no-lysine kinase 4)/SPAK (Ste20-related proline/alanine-rich kinase) which was blunted by NPR-C deletion. NCC inhibitor, hydrochlorothiazide, failed to induce natriuresis in NPR-C knockout mice. Moreover, low-salt and high-salt diets-induced changes of total and phosphorylation of NCC expression were normalized by NPR-C deletion. Importantly, tubule-specific deletion of NPR-C also attenuated Ang II-induced elevated blood pressure, total and phosphorylation of NCC expression. Mechanistically, in distal convoluted tubule cells, Ang II dose and time-dependently upregulated WNK4/SPAK/NCC kinase pathway and NPR-C/Gi/PLC/PKC signaling pathway mediated NCC activation. These results demonstrate that NPR-C signaling regulates NCC function contributing to sodium retention-mediated elevated blood pressure, which suggests that NPR-C is a promising candidate for the treatment of sodium retention-related hypertension.
Collapse
MESH Headings
- Angiotensin II
- Animals
- Blood Pressure/genetics
- Blood Pressure/physiology
- Cells, Cultured
- Female
- Hypertension/chemically induced
- Hypertension/genetics
- Hypertension/physiopathology
- Kidney/metabolism
- Kidney Tubules, Distal/cytology
- Kidney Tubules, Distal/metabolism
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Receptors, Atrial Natriuretic Factor/deficiency
- Receptors, Atrial Natriuretic Factor/genetics
- Renin-Angiotensin System/genetics
- Renin-Angiotensin System/physiology
- Signal Transduction/genetics
- Sodium/blood
- Sodium/urine
- Solute Carrier Family 12, Member 3/genetics
- Solute Carrier Family 12, Member 3/metabolism
- Mice
Collapse
Affiliation(s)
- Shuai Shao
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Xiao-Dong Li
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Yuan-Yuan Lu
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Shi-Jin Li
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Xiao-Hui Chen
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Han-Dan Zhou
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Shun He
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Yue-Tong Guo
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Xiao Lu
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Ping-Jin Gao
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| | - Ji-Guang Wang
- From the Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
265
|
Reynolds RJ, Irvin MR, Bridges SL, Kim H, Merriman TR, Arnett DK, Singh JA, Sumpter NA, Lupi AS, Vazquez AI. Genetic correlations between traits associated with hyperuricemia, gout, and comorbidities. Eur J Hum Genet 2021; 29:1438-1445. [PMID: 33637890 DOI: 10.1038/s41431-021-00830-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 12/06/2020] [Accepted: 02/10/2021] [Indexed: 01/26/2023] Open
Abstract
Hypertension, obesity, chronic kidney disease and type 2 diabetes are comorbidities that have very high prevalence among persons with hyperuricemia (serum urate > 6.8 mg/dL) and gout. Here we use multivariate genetic models to test the hypothesis that the co-association of traits representing hyperuricemia and its comorbidities is genetically based. Using Bayesian whole-genome regression models, we estimated the genetic marker-based variance and the covariance between serum urate, serum creatinine, systolic blood pressure (SBP), blood glucose and body mass index (BMI) from two independent family-based studies: The Framingham Heart Study-FHS and the Hypertension Genetic Epidemiology Network study-HyperGEN. The main genetic findings that replicated in both FHS and HyperGEN, were (1) creatinine was genetically correlated only with urate and (2) BMI was genetically correlated with urate, SBP, and glucose. The environmental covariance among the traits was generally highest for trait pairs involving BMI. The genetic overlap of traits representing the comorbidities of hyperuricemia and gout appears to cluster in two separate axes of genetic covariance. Because creatinine is genetically correlated with urate but not with metabolic traits, this suggests there is one genetic module of shared loci associated with hyperuricemia and chronic kidney disease. Another module of shared loci may account for the association of hyperuricemia and metabolic syndrome. This study provides a clear quantitative genetic basis for the clustering of comorbidities with hyperuricemia.
Collapse
Affiliation(s)
- Richard J Reynolds
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.
| | - M Ryan Irvin
- Department of Epidemiology, UAB, Birmingham, AL, USA
| | - S Louis Bridges
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Hwasoon Kim
- Duke Clinical Research Institute, Durham, NC, USA
| | - Tony R Merriman
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Donna K Arnett
- College of Public Health, University of Kentucky, Lexington, KY, USA
| | - Jasvinder A Singh
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA.,Birmingham VA Medical Center, Birmingham, AL, USA
| | - Nicholas A Sumpter
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham (UAB), Birmingham, AL, USA
| | - Alexa S Lupi
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA.,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA
| | - Ana I Vazquez
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA. .,Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
266
|
Li AL, Peng Q, Shao YQ, Fang X, Zhang YY. The interaction on hypertension between family history and diabetes and other risk factors. Sci Rep 2021; 11:4716. [PMID: 33633182 PMCID: PMC7907071 DOI: 10.1038/s41598-021-83589-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
To explore the individual effect and interaction of diabetes and family history and other risk factors on hypertension in Han in Shanghai China. The method of case-control study with l:l matched pairs was used, 342 cases of hypertension and 342 controls were selected and investigate their exposed factors with face-to-face. The method of epidemiology research was used to explore the individual effect and interaction of diabetes and family history and other risk factors on hypertension. The individual effect of family history (OR = 4.103, 95%CI 2.660-6.330), diabetes (OR = 4.219, 95%CI 2.926-6.083), personal taste (OR = 1.256, 95%CI 1.091-1.593), drinking behavior (OR = 1.391, 95%CI 1.010-1.914) and smoking behavior (OR = 1.057, 95%CI 1.00-1.117) were significant (p < 0.05). But individual effect of sex, education, occupation, work/life pressure, environmental noise, sleeping time and sports habit were not significant (p > 0.05). The OR of interaction between FH and DM to hypertension was 16.537 (95%CI 10.070-21.157), between FH and drinking behavior was 4.0 (95%CI 2.461-6.502), FH and sport habit was 7.668 (95%CI 3.598-16.344), FH and personal taste was 6.521 (95%CI 3.858-11.024), FH and smoking behavior was 5.526 (95%CI 3.404-8.972), FH and work/life pressure was 4.087 (95%CI 2.144-7.788). The SI of FH and DM was 2.27, RERI was 8.68, AP was 52.48% and PAP was 55.86%. FH and DM, personal taste, smoking behavior had positive interaction on hypertension, but FH and sport habits, drinking behavior, work/life pressure had reverse interaction on hypertension. FH and diabetes were very important risk factors with significant effect for hypertension. FH and diabetes, personal taste, smoking behavior had positive interaction on hypertension, but FH and sport habits, drinking behavior, work/life pressure had reverse interaction on hypertension.
Collapse
Affiliation(s)
- An-le Li
- Jiading District Center for Disease Control and Prevention, Shanghai, China.
| | - Qian Peng
- Jiading District Center for Disease Control and Prevention, Shanghai, China
| | - Yue-Qin Shao
- Jiading District Center for Disease Control and Prevention, Shanghai, China
| | - Xiang Fang
- Jiading District Center for Disease Control and Prevention, Shanghai, China
| | - Yi-Ying Zhang
- Jiading District Center for Disease Control and Prevention, Shanghai, China
| |
Collapse
|
267
|
Sandner P, Zimmer DP, Milne GT, Follmann M, Hobbs A, Stasch JP. Soluble Guanylate Cyclase Stimulators and Activators. Handb Exp Pharmacol 2021; 264:355-394. [PMID: 30689085 DOI: 10.1007/164_2018_197] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
When Furchgott, Murad, and Ignarro were honored with the Nobel prize for the identification of nitric oxide (NO) in 1998, the therapeutic implications of this discovery could not be fully anticipated. This was due to the fact that available therapeutics like NO donors did not allow a constant and long-lasting cyclic guanylyl monophosphate (cGMP) stimulation and had a narrow therapeutic window. Now, 20 years later, the stimulator of soluble guanylate cyclase (sGC), riociguat, is on the market and is the only drug approved for the treatment of two forms of pulmonary hypertension (PAH/CTEPH), and a variety of other sGC stimulators and sGC activators are in preclinical and clinical development for additional indications. The discovery of sGC stimulators and sGC activators is a milestone in the field of NO/sGC/cGMP pharmacology. The sGC stimulators and sGC activators bind directly to reduced, heme-containing and oxidized, heme-free sGC, respectively, which results in an increase in cGMP production. The action of sGC stimulators at the heme-containing enzyme is independent of NO but is enhanced in the presence of NO whereas the sGC activators interact with the heme-free form of sGC. These highly innovative pharmacological principles of sGC stimulation and activation seem to have a very broad therapeutic potential. Therefore, in both academia and industry, intensive research and development efforts have been undertaken to fully exploit the therapeutic benefit of these new compound classes. Here we summarize the discovery of sGC stimulators and sGC activators and the current developments in both compound classes, including the mode of action, the chemical structures, and the genesis of the terminology and nomenclature. In addition, preclinical studies exploring multiple aspects of their in vitro, ex vivo, and in vivo pharmacology are reviewed, providing an overview of multiple potential applications. Finally, the clinical developments, investigating the treatment potential of these compounds in various diseases like heart failure, diabetic kidney disease, fibrotic diseases, and hypertension, are reported. In summary, sGC stimulators and sGC activators have a unique mode of action with a broad treatment potential in cardiovascular diseases and beyond.
Collapse
Affiliation(s)
- Peter Sandner
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, Wuppertal, Germany. .,Department of Pharmacology, Hannover Medical School, Hannover, Germany.
| | | | | | - Markus Follmann
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, Wuppertal, Germany
| | - Adrian Hobbs
- Barts and the London School of Medicine and Dentistry QMUL, London, UK
| | - Johannes-Peter Stasch
- Bayer AG, Pharmaceuticals R&D, Pharma Research Center, Wuppertal, Germany.,Institute of Pharmacy, University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
268
|
Vaura F, Kauko A, Suvila K, Havulinna AS, Mars N, Salomaa V, FinnGen, Cheng S, Niiranen T. Polygenic Risk Scores Predict Hypertension Onset and Cardiovascular Risk. Hypertension 2021; 77:1119-1127. [PMID: 33611940 PMCID: PMC8025831 DOI: 10.1161/hypertensionaha.120.16471] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Felix Vaura
- From the Department of Internal Medicine, University of Turku, Finland (F.V., A.K., K.S., T.N.)
| | - Anni Kauko
- From the Department of Internal Medicine, University of Turku, Finland (F.V., A.K., K.S., T.N.)
| | - Karri Suvila
- From the Department of Internal Medicine, University of Turku, Finland (F.V., A.K., K.S., T.N.).,Division of Medicine, Turku University Hospital, Finland (K.S., T.N.)
| | - Aki S Havulinna
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland (A.S.H., V.S., T.N.).,Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki (A.S.H., N.M.)
| | - Nina Mars
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki (A.S.H., N.M.)
| | - Veikko Salomaa
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland (A.S.H., V.S., T.N.)
| | | | - Susan Cheng
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA (S.C.).,Division of Cardiology, Brigham and Women's Hospital, Boston, MA (S.C.)
| | - Teemu Niiranen
- From the Department of Internal Medicine, University of Turku, Finland (F.V., A.K., K.S., T.N.).,Division of Medicine, Turku University Hospital, Finland (K.S., T.N.).,Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland (A.S.H., V.S., T.N.)
| |
Collapse
|
269
|
Lei Y, Ludorf KL, Yu X, Benjamin RH, Gu X, Lin Y, Finnell RH, Mitchell LE, Musfee FI, Malik S, Canfield MA, Morrison AC, Hobbs CA, Van Zutphen AR, Fisher S, Agopian AJ. Maternal Hypertension-Related Genotypes and Congenital Heart Defects. Am J Hypertens 2021; 34:82-91. [PMID: 32710738 PMCID: PMC7891240 DOI: 10.1093/ajh/hpaa116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/05/2020] [Accepted: 07/20/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Maternal hypertension has been associated with congenital heart defect occurrence in several studies. We assessed whether maternal genotypes associated with this condition were also associated with congenital heart defect occurrence. METHODS We used data from the National Birth Defects Prevention Study to identify non-Hispanic white (NHW) and Hispanic women with (cases) and without (controls) a pregnancy in which a select simple, isolated heart defect was present between 1999 and 2011. We genotyped 29 hypertension-related single nucleotide polymorphisms (SNPs). We conducted logistic regression analyses separately by race/ethnicity to assess the relationship between the presence of any congenital heart defect and each SNP and an overall blood pressure genetic risk score (GRS). All analyses were then repeated to assess 4 separate congenital heart defect subtypes. RESULTS Four hypertension-related variants were associated with congenital heart defects among NHW women (N = 1,568 with affected pregnancies). For example, 1 intronic variant in ARHGAP2, rs633185, was associated with conotruncal defects (odds ratio [OR]: 1.3, 95% confidence interval [CI]: 1.1-1.6). Additionally, 2 variants were associated with congenital heart defects among Hispanic women (N = 489 with affected pregnancies). The GRS had a significant association with septal defects (OR: 2.1, 95% CI: 1.2-3.5) among NHW women. CONCLUSIONS We replicated a previously reported association between rs633185 and conotruncal defects. Although additional hypertension-related SNPs were also associated with congenital heart defects, more work is needed to better understand the relationship between genetic risk for maternal hypertension and congenital heart defects occurrence.
Collapse
Affiliation(s)
- Yunping Lei
- Department of Molecular and Cellular Biology and Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Katherine L Ludorf
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth School of Public Health, Houston, Texas, USA
| | - Xiao Yu
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth School of Public Health, Houston, Texas, USA
| | - Renata H Benjamin
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth School of Public Health, Houston, Texas, USA
| | - Xue Gu
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| | - Ying Lin
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| | - Richard H Finnell
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, Texas, USA
| | - Laura E Mitchell
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth School of Public Health, Houston, Texas, USA
| | - Fadi I Musfee
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth School of Public Health, Houston, Texas, USA
| | - Sadia Malik
- Pediatric Cardiology, Department of Pediatrics, UT Southwestern Children’s Medical Center, Dallas, Texas, USA
| | - Mark A Canfield
- Birth Defects Epidemiology and Surveillance Branch, Texas Department of State Health Services, Austin, Texas, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth School of Public Health, Houston, Texas, USA
| | - Charlotte A Hobbs
- Rady Children’s Institute for Genomic Medicine, San Diego, California, USA
| | - Alissa R Van Zutphen
- New York State Department of Health, Bureau of Environmental and Occupational Epidemiology, Albany, New York, USA
| | - Sarah Fisher
- New York State Department of Health, Bureau of Environmental and Occupational Epidemiology, Albany, New York, USA
| | - A J Agopian
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, UTHealth School of Public Health, Houston, Texas, USA
| |
Collapse
|
270
|
Dashti HS, Daghlas I, Lane JM, Huang Y, Udler MS, Wang H, Ollila HM, Jones SE, Kim J, Wood AR, Weedon MN, Aslibekyan S, Garaulet M, Saxena R. Genetic determinants of daytime napping and effects on cardiometabolic health. Nat Commun 2021; 12:900. [PMID: 33568662 PMCID: PMC7876146 DOI: 10.1038/s41467-020-20585-3] [Citation(s) in RCA: 136] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 12/08/2020] [Indexed: 12/14/2022] Open
Abstract
Daytime napping is a common, heritable behavior, but its genetic basis and causal relationship with cardiometabolic health remain unclear. Here, we perform a genome-wide association study of self-reported daytime napping in the UK Biobank (n = 452,633) and identify 123 loci of which 61 replicate in the 23andMe research cohort (n = 541,333). Findings include missense variants in established drug targets for sleep disorders (HCRTR1, HCRTR2), genes with roles in arousal (TRPC6, PNOC), and genes suggesting an obesity-hypersomnolence pathway (PNOC, PATJ). Association signals are concordant with accelerometer-measured daytime inactivity duration and 33 loci colocalize with loci for other sleep phenotypes. Cluster analysis identifies three distinct clusters of nap-promoting mechanisms with heterogeneous associations with cardiometabolic outcomes. Mendelian randomization shows potential causal links between more frequent daytime napping and higher blood pressure and waist circumference.
Collapse
Affiliation(s)
- Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Iyas Daghlas
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Jacqueline M Lane
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Miriam S Udler
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Heming Wang
- Broad Institute, Cambridge, MA, USA
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hanna M Ollila
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
- Institute for Molecular Medicine FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Samuel E Jones
- Institute for Molecular Medicine FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, UK
| | | | - Andrew R Wood
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, UK
| | - Michael N Weedon
- Genetics of Complex Traits, University of Exeter Medical School, Exeter, UK
| | | | - Marta Garaulet
- Division of Sleep and Circadian Disorders, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Physiology, University of Murcia, Murcia, Spain.
- IMIB-Arrixaca, Murcia, Spain.
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute, Cambridge, MA, USA.
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
271
|
Kuo CL, Joaquim M, Kuchel GA, Ferrucci L, Harries LW, Pilling LC, Melzer D. The Longevity-Associated SH2B3 (LNK) Genetic Variant: Selected Aging Phenotypes in 379,758 Subjects. J Gerontol A Biol Sci Med Sci 2021; 75:1656-1662. [PMID: 31428775 DOI: 10.1093/gerona/glz191] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Indexed: 12/15/2022] Open
Abstract
Human SH2B3 is involved in growth factor and inflammation signaling. A SH2B3 missense variant (rs3184504) is associated with cardiovascular diseases plus breast, colorectal, and lung cancers, with highly correlated variants across the ATXN2/SH2B3/BRAP locus linked to parental age at death, suggesting a geroscience common mechanism of aging and disease. To better understand the SH2B3-related aging pathway and its potential as an intervention target, we undertook a phenotype-wide association study (PheWAS) of 52 aging traits. Data were obtained from 379,758 European-descent UK Biobank participants, aged 40-70 at baseline: 27% of participants were CC homozygotes and 23% TT at rs3184504. Parental extreme longevity (mothers aged ≥98 years, fathers aged ≥96 years) was more common in CC versus TT (odds ratio [OR] = 1.18, 95% confidence interval [CI]: 1.07 to 1.29) with an additive per allele effect. The C allele associated with better cognitive function and white blood cell counts were more likely to be normal. The C allele reduced risks of coronary heart disease (OR = 0.95, 95% CI: 0.93 to 0.96) but was also associated with a modestly higher cancer rate (OR = 1.03, 95% CI: 1.02 to 1.04), suggesting a trade-off across aging outcomes and limiting its potential as an anti-aging target.
Collapse
Affiliation(s)
- Chia-Ling Kuo
- Connecticut Convergence Institute for Translation in Regenerative Engineering, University of Connecticut Health, Farmington
| | | | - George A Kuchel
- Center on Aging, University of Connecticut Health School of Medicine, Farmington
| | | | | | - Luke C Pilling
- College of Medicine and Health, University of Exeter, UK.,Center on Aging, University of Connecticut Health School of Medicine, Farmington
| | - David Melzer
- College of Medicine and Health, University of Exeter, UK.,Center on Aging, University of Connecticut Health School of Medicine, Farmington
| |
Collapse
|
272
|
Georgiopoulos G, Ntritsos G, Stamatelopoulos K, Tsioufis C, Aimo A, Masi S, Evangelou E. The relationship between blood pressure and risk of atrial fibrillation: a Mendelian randomization study. Eur J Prev Cardiol 2021; 29:zwab005. [PMID: 33556963 DOI: 10.1093/eurjpc/zwab005] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/12/2020] [Accepted: 01/06/2021] [Indexed: 01/07/2023]
Abstract
AIMS Observational studies suggest elevated blood pressure (BP) as the leading risk factor for incident atrial fibrillation (AF), but whether this relationship is causal remains unknown. In this study, we used Mendelian randomization (MR) to investigate the potential causal association of BP levels with the risk of developing AF. METHODS AND RESULTS Genetic variants associated with the BP traits were retrieved from the International Consortium of Blood Pressure-Genome Wide Association Studies (N = 299 024). From 901 reported variants, 894 were assessed in a dedicated Genome-Wide Association Study of AF genetics, including >1 000 000 subjects of European ancestry. We used two-sample MR analyses to examine the potential causal association of systolic BP (SBP) and diastolic BP (DBP) as well as of pulse pressure (PP) with AF. MR analysis identified a potentially causal association between AF and SBP [odds ratio (OR): 1.018 per 1 mmHg increase, 95% confidence interval (CI): 1.012-1.024, P < 0.001], DBP (OR: 1.026, 95% CI: 1.016-1.035, P < 0.001), and PP (OR: 1.014, 95% CI: 1.001-1.028, P = 0.033). These findings were robust in sensitivity analyses, including the MR-Egger method and the MR pleiotropy residual sum and outlier test (MR-PRESSO). The causal relationship of BP and AF did not change when single-nucleotide polymorphisms associated with possible confounders (i.e. coronary artery disease and obesity) of the causal relationship were excluded. CONCLUSIONS The association between increased BP levels and the risk of AF is likely causal and applies for different BP indices. Independently from other risk factors, optimal BP control might represent an important therapeutic target for AF prevention in the general population.
Collapse
Affiliation(s)
- Georgios Georgiopoulos
- School of Biomedical Engineering and Imaging Sciences, King's College London, Westminster Bridge Road, London SE1 7EH, UK
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Georgios Ntritsos
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Department of Informatics and Telecommunications, School of Informatics and Telecommunications, University of Ioannina, Arta, Greece
| | - Kimon Stamatelopoulos
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Costas Tsioufis
- First Department of Cardiology, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - Alberto Aimo
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy
- National Centre for Cardiovascular Prevention and Outcomes, Institute of Cardiovascular Science, University College London, London EC1A 4NP, UK
- Department of Twin Research & Genetic Epidemiology, King's College London, London WC2R 2LS, UK
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| |
Collapse
|
273
|
Liu L, Zeng P, Xue F, Yuan Z, Zhou X. Multi-trait transcriptome-wide association studies with probabilistic Mendelian randomization. Am J Hum Genet 2021; 108:240-256. [PMID: 33434493 PMCID: PMC7895847 DOI: 10.1016/j.ajhg.2020.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
A transcriptome-wide association study (TWAS) integrates data from genome-wide association studies and gene expression mapping studies for investigating the gene regulatory mechanisms underlying diseases. Existing TWAS methods are primarily univariate in nature, focusing on analyzing one outcome trait at a time. However, many complex traits are correlated with each other and share a common genetic basis. Consequently, analyzing multiple traits jointly through multivariate analysis can potentially improve the power of TWASs. Here, we develop a method, moPMR-Egger (multiple outcome probabilistic Mendelian randomization with Egger assumption), for analyzing multiple outcome traits in TWAS applications. moPMR-Egger examines one gene at a time, relies on its cis-SNPs that are in potential linkage disequilibrium with each other to serve as instrumental variables, and tests its causal effects on multiple traits jointly. A key feature of moPMR-Egger is its ability to test and control for potential horizontal pleiotropic effects from instruments, thus maximizing power while minimizing false associations for TWASs. In simulations, moPMR-Egger provides calibrated type I error control for both causal effects testing and horizontal pleiotropic effects testing and is more powerful than existing univariate TWAS approaches in detecting causal associations. We apply moPMR-Egger to analyze 11 traits from 5 trait categories in the UK Biobank. In the analysis, moPMR-Egger identified 13.15% more gene associations than univariate approaches across trait categories and revealed distinct regulatory mechanisms underlying systolic and diastolic blood pressures.
Collapse
Affiliation(s)
- Lu Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ping Zeng
- Department of Epidemiology and Biostatistics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
274
|
Foley CN, Staley JR, Breen PG, Sun BB, Kirk PDW, Burgess S, Howson JMM. A fast and efficient colocalization algorithm for identifying shared genetic risk factors across multiple traits. Nat Commun 2021; 12:764. [PMID: 33536417 PMCID: PMC7858636 DOI: 10.1038/s41467-020-20885-8] [Citation(s) in RCA: 255] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 12/17/2020] [Indexed: 01/30/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified thousands of genomic regions affecting complex diseases. The next challenge is to elucidate the causal genes and mechanisms involved. One approach is to use statistical colocalization to assess shared genetic aetiology across multiple related traits (e.g. molecular traits, metabolic pathways and complex diseases) to identify causal pathways, prioritize causal variants and evaluate pleiotropy. We propose HyPrColoc (Hypothesis Prioritisation for multi-trait Colocalization), an efficient deterministic Bayesian algorithm using GWAS summary statistics that can detect colocalization across vast numbers of traits simultaneously (e.g. 100 traits can be jointly analysed in around 1 s). We perform a genome-wide multi-trait colocalization analysis of coronary heart disease (CHD) and fourteen related traits, identifying 43 regions in which CHD colocalized with ≥1 trait, including 5 previously unknown CHD loci. Across the 43 loci, we further integrate gene and protein expression quantitative trait loci to identify candidate causal genes.
Collapse
Affiliation(s)
- Christopher N Foley
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, CB2 0SR, UK.
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK.
| | - James R Staley
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
- MRC Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Philip G Breen
- School of Mathematics, University of Edinburgh, Kings Buildings, Edinburgh, EH9 3JZ, UK
| | - Benjamin B Sun
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Paul D W Kirk
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, CB2 0SR, UK
| | - Stephen Burgess
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, CB2 0SR, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Joanna M M Howson
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
- National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, UK
- Department of Genetics, Novo Nordisk Research Centre Oxford, Oxford, UK
| |
Collapse
|
275
|
The electrogenic sodium bicarbonate cotransporter and its roles in the myocardial ischemia-reperfusion induced cardiac diseases. Life Sci 2021; 270:119153. [PMID: 33539911 DOI: 10.1016/j.lfs.2021.119153] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/06/2021] [Accepted: 01/22/2021] [Indexed: 12/19/2022]
Abstract
Cardiac tissue ischemia/hypoxia increases glycolysis and lactic acid accumulation in cardiomyocytes, leading to intracellular metabolic acidosis. Sodium bicarbonate cotransporters (NBCs) play a vital role in modulating intracellular pH and maintaining sodium ion concentrations in cardiomyocytes. Cardiomyocytes mainly express electrogenic sodium bicarbonate cotransporter (NBCe1), which has been demonstrated to participate in myocardial ischemia/reperfusion (I/R) injury. This review outlines the structural and functional properties of NBCe1, summarizes the signaling pathways and factors that may regulate the activity of NBCe1, and reviews the roles of NBCe1 in the pathogenesis of I/R-induced cardiac diseases. Further studies revealing the regulatory mechanisms of NBCe1 activity should provide novel therapeutic targets for preventing I/R-induced cardiac diseases.
Collapse
|
276
|
Catov JM, McNeil RB, Marsh DJ, Mercer BM, Bairey Merz CN, Parker CB, Pemberton VL, Saade GR, Chen Y(I, Chung JH, Ehrenthal DB, Grobman WA, Haas DM, Parry S, Polito L, Reddy UM, Silver RM, Simhan HN, Wapner RJ, Kominiarek M, Kreutz R, Levine LD, Greenland P, for the NHLBI nuMoM2b Heart Health Study. Early Pregnancy Atherogenic Profile in a First Pregnancy and Hypertension Risk 2 to 7 Years After Delivery. J Am Heart Assoc 2021; 10:e017216. [PMID: 33619977 PMCID: PMC8174276 DOI: 10.1161/jaha.120.017216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022]
Abstract
Background Cardiovascular risk in young adulthood is an important determinant of lifetime cardiovascular disease risk. Women with adverse pregnancy outcomes (APOs) have increased cardiovascular risk, but the relationship of other factors is unknown. Methods and Results Among 4471 primiparous women, we related first-trimester atherogenic markers to risk of APO (hypertensive disorders of pregnancy, preterm birth, small for gestational age), gestational diabetes mellitus (GDM) and hypertension (130/80 mm Hg or antihypertensive use) 2 to 7 years after delivery. Women with an APO/GDM (n=1102) had more atherogenic characteristics (obesity [34.2 versus 19.5%], higher blood pressure [systolic blood pressure 112.2 versus 108.4, diastolic blood pressure 69.2 versus 66.6 mm Hg], glucose [5.0 versus 4.8 mmol/L], insulin [77.6 versus 60.1 pmol/L], triglycerides [1.4 versus 1.3 mmol/L], and high-sensitivity C-reactive protein [5.6 versus 4.0 nmol/L], and lower high-density lipoprotein cholesterol [1.8 versus 1.9 mmol/L]; P<0.05) than women without an APO/GDM. They were also more likely to develop hypertension after delivery (32.8% versus 18.1%, P<0.05). Accounting for confounders and factors routinely assessed antepartum, higher glucose (relative risk [RR] 1.03 [95% CI, 1.00-1.06] per 0.6 mmol/L), high-sensitivity C-reactive protein (RR, 1.06 [95% CI, 1.02-1.11] per 2-fold higher), and triglycerides (RR, 1.27 [95% CI, 1.14-1.41] per 2-fold higher) were associated with later hypertension. Higher physical activity was protective (RR, 0.93 [95% CI, 0.87-0.99] per 3 h/week). When evaluated as latent profiles, the nonobese group with higher lipids, high-sensitivity C-reactive protein, and insulin values (6.9% of the cohort) had increased risk of an APO/GDM and later hypertension. Among these factors, 7% to 15% of excess RR was related to APO/GDM. Conclusions Individual and combined first-trimester atherogenic characteristics are associated with APO/GDM occurrence and hypertension 2 to 7 years later. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT02231398.
Collapse
Affiliation(s)
| | | | | | - Brian M. Mercer
- Case Western Reserve University–The MetroHealth SystemClevelandOH
| | | | | | | | | | | | | | | | | | | | - Samuel Parry
- University of Pennsylvania School of MedicinePhiladelphiaPA
| | - LuAnn Polito
- Case Western Reserve University–The MetroHealth SystemClevelandOH
| | - Uma M. Reddy
- Eunice Kennedy Shriver National Institute of Child Health and Human DevelopmentBethesdaMD
| | | | | | | | | | - Rolf Kreutz
- Indiana University School of MedicineIndianapolisIN
| | - Lisa D. Levine
- University of Pennsylvania School of MedicinePhiladelphiaPA
| | | | | |
Collapse
|
277
|
Hyman MC, Levin MG, Gill D, Walker VM, Georgakis MK, Davies NM, Marchlinski FE, Damrauer SM. Genetically Predicted Blood Pressure and Risk of Atrial Fibrillation. Hypertension 2021; 77:376-382. [PMID: 33390040 PMCID: PMC7803440 DOI: 10.1161/hypertensionaha.120.16191] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/18/2020] [Indexed: 01/02/2023]
Abstract
Observational studies have shown an association between hypertension and atrial fibrillation (AF). Aggressive blood pressure management in patients with known AF reduces overall arrhythmia burden, but it remains unclear whether hypertension is causative for AF. To address this question, this study explored the relationship between genetic predictors of blood pressure and risk of AF. We secondarily explored the relationship between genetically proxied use of antihypertensive drugs and risk of AF. Two-sample Mendelian randomization was performed using an inverse-variance weighted meta-analysis with weighted median Mendelian randomization and Egger intercept tests performed as sensitivity analyses. Summary statistics for systolic blood pressure, diastolic blood pressure, and pulse pressure were obtained from the International Consortium of Blood Pressure and the UK Biobank discovery analysis and AF from the 2018 Atrial Fibrillation Genetics Consortium multiethnic genome-wide association studies. Increases in genetically proxied systolic blood pressure, diastolic blood pressure, or pulse pressure by 10 mm Hg were associated with increased odds of AF (systolic blood pressure: odds ratio [OR], 1.17 [95% CI, 1.11-1.22]; P=1×10-11; diastolic blood pressure: OR, 1.25 [95% CI, 1.16-1.35]; P=3×10-8; pulse pressure: OR, 1.1 [95% CI, 1.0-1.2]; P=0.05). Decreases in systolic blood pressure by 10 mm Hg estimated by genetic proxies of antihypertensive medications showed calcium channel blockers (OR, 0.66 [95% CI, 0.57-0.76]; P=8×10-9) and β-blockers (OR, 0.61 [95% CI, 0.46-0.81]; P=6×10-4) decreased the risk of AF. Blood pressure-increasing genetic variants were associated with increased risk of AF, consistent with a causal relationship between blood pressure and AF. These data support the concept that blood pressure reduction with calcium channel blockade or β-blockade could reduce the risk of AF.
Collapse
Affiliation(s)
- Matthew C. Hyman
- From the Division of Cardiovascular Medicine (M.C.H., M.G.L., F.E.M.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Michael G. Levin
- From the Division of Cardiovascular Medicine (M.C.H., M.G.L., F.E.M.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA (M.G.L., S.M.D.)
| | - Dipender Gill
- Department of Epidemiology and Biostatistics, School of Public Health (D.G.), Imperial College London, United Kingdom
- Department of Medicine, Centre for Pharmacology and Therapeutics, Hammersmith Campus (D.G.), Imperial College London, United Kingdom
- Department of Genetics, Novo Nordisk Research Centre Oxford, Old Road Campus, United Kingdom (D.G.)
- Clinical Pharmacology and Therapeutics Section, Institute of Medical and Biomedical Education and Institute for Infection and Immunity, St George’s, University of London, United Kingdom (D.G.)
- Clinical Pharmacology Group, Pharmacy and Medicines Directorate, St George’s University Hospitals NHS Foundation Trust, London, United Kingdom (D.G.)
| | - Venexia M. Walker
- Department of Surgery (V.M.W., S.M.D.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Medical Research Council Integrative Epidemiology Unit (V.M.W., N.D.), University of Bristol, United Kingdom
- Bristol Medical School: Population Health Sciences (V.M.W.), University of Bristol, United Kingdom
| | - Marios K. Georgakis
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-University LMU, Munich, Germany (M.K.G.)
| | - Neil M. Davies
- Medical Research Council Integrative Epidemiology Unit (V.M.W., N.D.), University of Bristol, United Kingdom
| | - Francis E. Marchlinski
- From the Division of Cardiovascular Medicine (M.C.H., M.G.L., F.E.M.), University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Scott M. Damrauer
- Department of Surgery (V.M.W., S.M.D.), University of Pennsylvania Perelman School of Medicine, Philadelphia
- Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA (M.G.L., S.M.D.)
| |
Collapse
|
278
|
Silva CP, Kamens HM. Cigarette smoke-induced alterations in blood: A review of research on DNA methylation and gene expression. Exp Clin Psychopharmacol 2021; 29:116-135. [PMID: 32658533 PMCID: PMC7854868 DOI: 10.1037/pha0000382] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Worldwide, smoking remains a threat to public health, causing preventable diseases and premature mortality. Cigarette smoke is a powerful inducer of DNA methylation and gene expression alterations, which have been associated with negative health consequences. Here, we review the current knowledge on smoking-related changes in DNA methylation and gene expression in human blood samples. We identified 30 studies focused on the association between active smoking, DNA methylation modifications, and gene expression alterations. Overall, we identified 1,758 genes with differentially methylated sites (DMS) and differentially expressed genes (DEG) between smokers and nonsmokers, of which 261 were detected in multiple studies (≥4). The most frequently (≥10 studies) reported genes were AHRR, GPR15, GFI1, and RARA. Functional enrichment analysis of the 261 genes identified the aryl hydrocarbon receptor repressor and T cell pathways (T helpers 1 and 2) as influenced by smoking status. These results highlight specific genes for future mechanistic and translational research that may be associated with cigarette smoke exposure and smoking-related diseases. (PsycInfo Database Record (c) 2021 APA, all rights reserved).
Collapse
Affiliation(s)
- Constanza P. Silva
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America
| | - Helen M. Kamens
- Department of Biobehavioral Health, The Pennsylvania State University, University Park, Pennsylvania, 16802, United States of America.,Correspondence concerning this article should be addressed to Helen M. Kamens, 228 Biobehavioral Health Building, The Pennsylvania State University, University Park, PA 16802; ; Phone number: 814-865-1269; Fax number: 814-863-7525
| |
Collapse
|
279
|
Park JM, Park DH, Song Y, Kim JO, Choi JE, Kwon YJ, Kim SJ, Lee JW, Hong KW. Understanding the genetic architecture of the metabolically unhealthy normal weight and metabolically healthy obese phenotypes in a Korean population. Sci Rep 2021; 11:2279. [PMID: 33500527 PMCID: PMC7838176 DOI: 10.1038/s41598-021-81940-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/14/2021] [Indexed: 01/30/2023] Open
Abstract
Understanding the mechanisms underlying the metabolically unhealthy normal weight (MUHNW) and metabolically healthy obese (MHO) phenotypes is important for developing strategies to prevent cardiometabolic diseases. Here, we conducted genome-wide association studies (GWASs) to identify the MUHNW and MHO genetic indices. The study dataset comprised genome-wide single-nucleotide polymorphism genotypes and epidemiological data from 49,915 subjects categorised into four phenotypes-metabolically healthy normal weight (MHNW), MUHNW, MHO, and metabolically unhealthy obese (MUHO). We conducted two GWASs using logistic regression analyses and adjustments for confounding variables (model 1: MHNW versus MUHNW and model 2: MHO versus MUHO). GCKR, ABCB11, CDKAL1, LPL, CDKN2B, NT5C2, APOA5, CETP, and APOC1 were associated with metabolically unhealthy phenotypes among normal weight individuals (model 1). LPL, APOA5, and CETP were associated with metabolically unhealthy phenotypes among obese individuals (model 2). The genes common to both models are related to lipid metabolism (LPL, APOA5, and CETP), and those associated with model 1 are related to insulin or glucose metabolism (GCKR, CDKAL1, and CDKN2B). This study reveals the genetic architecture of the MUHNW and MHO phenotypes in a Korean population-based cohort. These findings could help identify individuals at a high metabolic risk in normal weight and obese populations and provide potential novel targets for the management of metabolically unhealthy phenotypes.
Collapse
Affiliation(s)
- Jae-Min Park
- grid.15444.300000 0004 0470 5454Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju‐ro, Gangnam-gu, Seoul, 06273 Korea ,grid.15444.300000 0004 0470 5454Department of Medicine, Graduate School of Medicine, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722 Korea
| | - Da-Hyun Park
- Theragen Bio Co., Ltd., 145 Gwanggyo-ro, Suwon-si, Gyeonggi-do 16229 Korea
| | - Youhyun Song
- grid.15444.300000 0004 0470 5454Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju‐ro, Gangnam-gu, Seoul, 06273 Korea
| | - Jung Oh Kim
- Theragen Bio Co., Ltd., 145 Gwanggyo-ro, Suwon-si, Gyeonggi-do 16229 Korea
| | - Ja-Eun Choi
- Theragen Bio Co., Ltd., 145 Gwanggyo-ro, Suwon-si, Gyeonggi-do 16229 Korea
| | - Yu-Jin Kwon
- grid.15444.300000 0004 0470 5454Department of Family Medicine, Yongin Severance Hospital, Yonsei University College of Medicine, 363 Dongbaekjukjeon-daero, Giheung-gu, Yongin-si, Gyeonggi-do 16995 Korea
| | - Seong-Jin Kim
- Theragen Bio Co., Ltd., 145 Gwanggyo-ro, Suwon-si, Gyeonggi-do 16229 Korea
| | - Ji-Won Lee
- grid.15444.300000 0004 0470 5454Department of Family Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju‐ro, Gangnam-gu, Seoul, 06273 Korea
| | - Kyung-Won Hong
- Theragen Bio Co., Ltd., 145 Gwanggyo-ro, Suwon-si, Gyeonggi-do 16229 Korea
| |
Collapse
|
280
|
Kahali B, Chen Y, Feitosa MF, Bielak LF, O’Connell JR, Musani SK, Hegde Y, Chen Y, Stetson LC, Guo X, Fu YP, Smith AV, Ryan KA, Eiriksdottir G, Cohain AT, Allison M, Bakshi A, Bowden DW, Budoff MJ, Carr JJ, Carskadon S, Chen YDI, Correa A, Crudup BF, Du X, Harris TB, Yang J, Kardia SLR, Launer LJ, Liu J, Mosley TH, Norris JM, Terry JG, Palanisamy N, Schadt EE, O’Donnell CJ, Yerges-Armstrong LM, Rotter JI, Wagenknecht LE, Handelman SK, Gudnason V, Province MA, Peyser PA, Halligan B, Palmer ND, Speliotes EK. A Noncoding Variant Near PPP1R3B Promotes Liver Glycogen Storage and MetS, but Protects Against Myocardial Infarction. J Clin Endocrinol Metab 2021; 106:372-387. [PMID: 33231259 PMCID: PMC7823249 DOI: 10.1210/clinem/dgaa855] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Indexed: 01/02/2023]
Abstract
CONTEXT Glycogen storage diseases are rare. Increased glycogen in the liver results in increased attenuation. OBJECTIVE Investigate the association and function of a noncoding region associated with liver attenuation but not histologic nonalcoholic fatty liver disease. DESIGN Genetics of Obesity-associated Liver Disease Consortium. SETTING Population-based. MAIN OUTCOME Computed tomography measured liver attenuation. RESULTS Carriers of rs4841132-A (frequency 2%-19%) do not show increased hepatic steatosis; they have increased liver attenuation indicative of increased glycogen deposition. rs4841132 falls in a noncoding RNA LOC157273 ~190 kb upstream of PPP1R3B. We demonstrate that rs4841132-A increases PPP1R3B through a cis genetic effect. Using CRISPR/Cas9 we engineered a 105-bp deletion including rs4841132-A in human hepatocarcinoma cells that increases PPP1R3B, decreases LOC157273, and increases glycogen perfectly mirroring the human disease. Overexpression of PPP1R3B or knockdown of LOC157273 increased glycogen but did not result in decreased LOC157273 or increased PPP1R3B, respectively, suggesting that the effects may not all occur via affecting RNA levels. Based on electronic health record (EHR) data, rs4841132-A associates with all components of the metabolic syndrome (MetS). However, rs4841132-A associated with decreased low-density lipoprotein (LDL) cholesterol and risk for myocardial infarction (MI). A metabolic signature for rs4841132-A includes increased glycine, lactate, triglycerides, and decreased acetoacetate and beta-hydroxybutyrate. CONCLUSIONS These results show that rs4841132-A promotes a hepatic glycogen storage disease by increasing PPP1R3B and decreasing LOC157273. rs4841132-A promotes glycogen accumulation and development of MetS but lowers LDL cholesterol and risk for MI. These results suggest that elevated hepatic glycogen is one cause of MetS that does not invariably promote MI.
Collapse
Affiliation(s)
- Bratati Kahali
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Centre for Brain Research, Indian Institute of Science, Bangalore, India
| | - Yue Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mary F Feitosa
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Lawrence F Bielak
- School of Public Health, Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Jeffrey R O’Connell
- Department of Endocrinology, Diabetes, and Nutrition, University of Maryland-Baltimore, Baltimore, MD, USA
| | - Solomon K Musani
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yash Hegde
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Yanhua Chen
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - L C Stetson
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, LABioMed and Department of Pediatrics at Harbor-UCLA, Torrance, CA, USA
| | - Yi-ping Fu
- Framingham Heart Study, NHLBI, NIH, Framingham, MA, USA
- Office of Biostatistics Research, Division of Cardiovascular Diseases, NHLBI, NIH, Bethesda, MD, USA
| | - Albert Vernon Smith
- School of Public Health, Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Kathleen A Ryan
- Department of Endocrinology, Diabetes, and Nutrition, University of Maryland-Baltimore, Baltimore, MD, USA
| | | | - Ariella T Cohain
- Department of Genetics and Genomics Sciences, Icahn School of Medicine, New York, NY, USA
| | - Matthew Allison
- Department of Family Medicine and Public Health, University of California, San Diego, CA, USA
| | - Andrew Bakshi
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Matthew J Budoff
- Department of Internal Medicine, LA Biomedical Research Institute at Harbor-UCLA, Torrance, CA, USA
| | - J Jeffrey Carr
- Department of Radiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Yii-Der I Chen
- Institute for Translational Genomics and Population Sciences, LABioMed and Department of Pediatrics at Harbor-UCLA, Torrance, CA, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Breland F Crudup
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xiaomeng Du
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Tamara B Harris
- Laboratory of Epidemiology and Population Sciences, National Institute of Aging, Bethesda, MD, USA
| | - Jian Yang
- Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Sharon L R Kardia
- School of Public Health, Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute of Aging, Bethesda, MD, USA
| | - Jiankang Liu
- Brigham and Women’s Hospital, Havard University, Boston, MA, USA
| | - Thomas H Mosley
- Department of Medicine, Division of Geriatrics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jill M Norris
- Department of Preventive Medicine and Biometrics, University of Colorado at Denver Health Sciences Center, Aurora, CO, USA
| | - James G Terry
- Department of Radiology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Eric E Schadt
- Department of Genetics and Genomics Sciences, Icahn School of Medicine, New York, NY, USA
| | - Christopher J O’Donnell
- Framingham Heart Study, NHLBI, NIH, Framingham, MA, USA
- Cardiology Section, Department of Medicine, Boston Veteran’s Administration Healthcare, Boston, MA, USA
| | - Laura M Yerges-Armstrong
- Department of Endocrinology, Diabetes, and Nutrition, University of Maryland-Baltimore, Baltimore, MD, USA
- Target Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, LABioMed and Department of Pediatrics at Harbor-UCLA, Torrance, CA, USA
| | - Lynne E Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Samuel K Handelman
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur, Iceland
- Department of Medicine, University of Iceland, Reykjavik, Iceland
| | - Michael A Province
- Division of Statistical Genomics, Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Patricia A Peyser
- School of Public Health, Department of Epidemiology, University of Michigan, Ann Arbor, MI, USA
| | - Brian Halligan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Elizabeth K Speliotes
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
281
|
Li Y, Zhang Q, Di Zhang, Cai Q, Fan J, Venners SA, Jiang S, Li J, Xu X. The effect of ABCA1 gene DNA methylation on blood pressure levels in a Chinese hyperlipidemic population. J Hum Hypertens 2021; 35:1139-1148. [PMID: 33462393 DOI: 10.1038/s41371-020-00479-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 06/08/2020] [Accepted: 12/11/2020] [Indexed: 11/09/2022]
Abstract
Hypertension is an important public health challenge worldwide. Epigenetic studies are providing novel insight into the underlying mechanisms of hypertension. We investigated the effect of DNA methylation in ATP-binding cassette transporter 1 (ABCA1) gene on blood pressure levels in a Chinese hyperlipidemic population. We randomly selected 211 individuals with hyperlipidemia who had not received any lipid-lowering treatment at baseline from our previous statin pharmacogenetics study (n = 734). DNA methylation loci at the ABCA1 gene were measured by MethylTarget, a next generation bisulfite sequencing-based multiple targeted cytosine-guanine dinucleotide methylation analysis method. Mean DNA methylation level was used in statistical analysis. In all subjects, higher mean ABCA1_B methylation was positively associated with systolic blood pressure (SBP) (β = 8.27, P = 0.008; β = 8.78, P = 0.005) and explained 2.7% and 5.8% of SBP variation before and after adjustment for lipids, respectively. We further divided all patients into three groups based on the tertile of body mass index (BMI) distribution. In the middle tertile of BMI, there was a significantly positive relationship between mean ABCA1_A methylation and SBP (β = 0.89, P = 0.003) and DBP (β = 0.32, P = 0.030). Mean ABCA1_A methylation explained 11.0% of SBP variation and 5.3% of DBP variation, respectively. Furthermore, mean ABCA1_A methylation (β = 0.79; P = 0.007) together with age and gender explained up to 24.1% of SBP variation. Our study provides new evidence that the ABCA1 DNA methylation profile is associated with blood pressure levels, which highlights that DNA methylation might be a significant molecular mechanism involved in the pathophysiological process of hypertension.
Collapse
Affiliation(s)
- Yajie Li
- School of Life Sciences, Anhui University, Hefei, China
| | - Qian Zhang
- School of Life Sciences, Anhui University, Hefei, China
| | - Di Zhang
- School of Life Sciences, Anhui University, Hefei, China
| | - Qianru Cai
- School of Life Sciences, Anhui University, Hefei, China
| | - Juanlin Fan
- School of Life Sciences, Anhui University, Hefei, China
| | - Scott A Venners
- Faculty of Health Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Shanqun Jiang
- School of Life Sciences, Anhui University, Hefei, China. .,Institute of Biomedicine, Anhui Medical University, Hefei, China.
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China.
| | - Xiping Xu
- Institute of Biomedicine, Anhui Medical University, Hefei, China.,National Clinical Research Study Center for Kidney Disease, State Key Laboratory for Organ Failure Research, Renal Division, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
282
|
Association of Age of Onset of Hypertension With Cardiovascular Diseases and Mortality. J Am Coll Cardiol 2021; 75:2921-2930. [PMID: 32527401 DOI: 10.1016/j.jacc.2020.04.038] [Citation(s) in RCA: 268] [Impact Index Per Article: 67.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 04/14/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND The relations of hypertension onset age with cardiovascular diseases (CVD) and all-cause mortality remain inconclusive. OBJECTIVES This study sought to examine the associations of hypertension onset age with CVD and all-cause mortality. METHODS This prospective study included 71,245 participants free of hypertension and CVD in the first survey (July 2006 to October 2007) of the Kailuan study, a prospective cohort study in Tangshan, China. All participants were followed biennially until December 31, 2017. A total of 20,221 new-onset hypertension cases were identified during follow-up. We randomly selected 1 control participant for each new-onset hypertensive participant, matching for age (±1 year) and sex, and included 19,887 case-control pairs. We used weighted Cox regression models to calculate the average hazard ratios of incident CVD and all-cause mortality across the age groups. RESULTS During an average follow-up of 6.5 years, we identified 1,672 incident CVD cases and 2,008 deaths. After multivariate adjustment, with the increase in hypertension onset age, the hazards of outcomes were gradually attenuated. The average hazard ratio (95% confidence interval) of CVD and all-cause mortality were 2.26 (1.19 to 4.30) and 2.59 (1.32 to 5.07) for the hypertension onset age <45 years old group, 1.62 (1.24 to 2.12) and 2.12 (1.55 to 2.90) for the 45- to 54-year age group, 1.42 (1.12 to 1.79) and 1.30 (1.03 to 1.62) for the 55- to 64-year age group, and 1.33 (1.04 to 1.69) and 1.29 (1.11 to 1.51) for the ≥65-year age group, respectively (p for interaction = 0.38 for CVD and <0.01 for death). CONCLUSIONS Hypertension was associated with a higher risk for CVD and all-cause mortality, and the associations were stronger with a younger age of onset.
Collapse
|
283
|
Laaksonen J, Mishra PP, Seppälä I, Lyytikäinen LP, Raitoharju E, Mononen N, Lepistö M, Almusa H, Ellonen P, Hutri-Kähönen N, Juonala M, Raitakari O, Kähönen M, Salonen JT, Lehtimäki T. Examining the effect of mitochondrial DNA variants on blood pressure in two Finnish cohorts. Sci Rep 2021; 11:611. [PMID: 33436758 PMCID: PMC7804469 DOI: 10.1038/s41598-020-79931-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022] Open
Abstract
High blood pressure (BP) is a major risk factor for many noncommunicable diseases. The effect of mitochondrial DNA single-nucleotide polymorphisms (mtSNPs) on BP is less known than that of nuclear SNPs. We investigated the mitochondrial genetic determinants of systolic, diastolic, and mean arterial BP. MtSNPs were determined from peripheral blood by sequencing or with genome-wide association study SNP arrays in two independent Finnish cohorts, the Young Finns Study and the Finnish Cardiovascular Study, respectively. In total, over 4200 individuals were included. The effects of individual common mtSNPs, with an additional focus on sex-specificity, and aggregates of rare mtSNPs grouped by mitochondrial genes were evaluated by meta-analysis of linear regression and a sequence kernel association test, respectively. We accounted for the predicted pathogenicity of the rare variants within protein-encoding and the tRNA regions. In the meta-analysis of 87 common mtSNPs, we did not observe significant associations with any of the BP traits. Sex-specific and rare-variant analyses did not pinpoint any significant associations either. Our results are in agreement with several previous studies suggesting that mtDNA variation does not have a significant role in the regulation of BP. Future studies might need to reconsider the mechanisms thought to link mtDNA with hypertension.
Collapse
Affiliation(s)
- Jaakko Laaksonen
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland.
| | - Pashupati P Mishra
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| | - Maija Lepistö
- Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland
| | - Henrikki Almusa
- Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland
| | - Pekka Ellonen
- Institute for Molecular Medicine (FIMM), University of Helsinki, Helsinki, Finland
| | - Nina Hutri-Kähönen
- Department of Paediatrics, Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Markus Juonala
- Department of Medicine, University of Turku, Turku, Finland.,Division of Medicine, Turku University Hospital, Turku, Finland.,Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Olli Raitakari
- Centre for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland.,Research Centre for Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland.,Department of Clinical Physiology and Nuclear Medicine, University of Turku and Turku University Hospital, Turku, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jukka T Salonen
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,MAS-Metabolic Analytical Services Oy, Helsinki, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, PO Box 100, 33014, Tampere, Finland
| |
Collapse
|
284
|
Abstract
Cyclic guanosine 3',5'-monophosphate (cGMP) is the key second messenger molecule in nitric oxide signaling. Its rapid generation and fate, but also its role in mediating acute cellular functions has been extensively studied. In the past years, genetic studies suggested an important role for cGMP in affecting the risk of chronic cardiovascular diseases, for example, coronary artery disease and myocardial infarction. Here, we review the role of cGMP in atherosclerosis and other cardiovascular diseases and discuss recent genetic findings and identified mechanisms. Finally, we highlight open questions and promising research topics.
Collapse
|
285
|
Zhou L, Wen X, Peng Y, Zhao L, Yu Y. Salt added to food and body mass index: A bidirectional Mendelian randomisation study. Nutr Diet 2021; 78:315-323. [PMID: 33415802 DOI: 10.1111/1747-0080.12653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 11/28/2022]
Abstract
AIM This study aimed to determine the causal association between salt added to food and body mass index (BMI) by integrating a summary-level genome-wide association study (GWAS) data. METHODS We performed two-sample Mendelian randomisation (MR) analyses using summary statistics of GWAS. Inverse-variance weighted (IVW), maximum likelihood estimation, and random effect model were used to analyse the effect of salt added to food on BMI. A bidirectional MR analysis with BMI as the exposure and salt added to food as the outcome was also performed. RESULTS The single nucleotide polymorphisms (SNPs) were selected from the UK Biobank (n = 462 630) and a meta-analysis of 322 154 European-descent individuals. The IVW method estimate indicated that salt added to food was positively associated with BMI (β = 0.1416, SE = 0.0576, P = .0139). Results from maximum likelihood estimation (β = 0.1476, SE = 0.0363, P < .0001) and the random effect model (β = 0.1411, SE = 0.0572, P = .0137) were consistent with the IVW. Bidirectional MR analysis suggested that BMI did not causally affect salt added to food. CONCLUSION Our results provided qualitative evidence supporting a causal relationship between salt intake and BMI.
Collapse
Affiliation(s)
- Long Zhou
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiaoxiao Wen
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaguang Peng
- Center for Clinical Epidemiology and Evidence-based Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing, China
| | - Liancheng Zhao
- Division of Prevention and Community Health, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Yu
- School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, China
| |
Collapse
|
286
|
Chatterjee S, Ouidir M, Tekola-Ayele F. Pleiotropic genetic influence on birth weight and childhood obesity. Sci Rep 2021; 11:48. [PMID: 33420178 PMCID: PMC7794220 DOI: 10.1038/s41598-020-80084-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 12/09/2020] [Indexed: 01/09/2023] Open
Abstract
Childhood obesity is a global public health problem. Understanding the molecular mechanisms that underlie early origins of childhood obesity can facilitate interventions. Consistent phenotypic and genetic correlations have been found between childhood obesity traits and birth weight (a proxy for in-utero growth), suggesting shared genetic influences (pleiotropy). We aimed to (1) investigate whether there is significant shared genetic influence between birth weight and childhood obesity traits, and (2) to identify genetic loci with shared effects. Using a statistical approach that integrates summary statistics and functional annotations for paired traits, we found strong evidence of pleiotropy (P < 3.53 × 10–127) and enrichment of functional annotations (P < 1.62 × 10–39) between birth weight and childhood body mass index (BMI)/obesity. The pleiotropic loci were enriched for regulatory features in skeletal muscle, adipose and brain tissues and in cell lines derived from blood lymphocytes. At 5% false discovery rate, 6 loci were associated with birth weight and childhood BMI and 13 loci were associated with birth weight and childhood obesity. Out of these 19 loci, one locus (EBF1) was novel to childhood obesity and one locus (LMBR1L) was novel to both birth weight and childhood BMI/obesity. These findings give evidence of substantial shared genetic effects in the regulation of both fetal growth and childhood obesity.
Collapse
Affiliation(s)
- Suvo Chatterjee
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, 20892-7004, USA
| | - Marion Ouidir
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, 20892-7004, USA
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, 6710B Rockledge Drive, Room 3204, Bethesda, 20892-7004, USA.
| |
Collapse
|
287
|
Polygenic overlap and shared genetic loci between loneliness, severe mental disorders, and cardiovascular disease risk factors suggest shared molecular mechanisms. Transl Psychiatry 2021; 11:3. [PMID: 33414458 PMCID: PMC7790035 DOI: 10.1038/s41398-020-01142-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 11/25/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
Clinical and epidemiological evidence suggest that loneliness is associated with severe mental disorders (SMDs) and increases the risk of cardiovascular disease (CVD). However, the mechanisms underlying the relationship between loneliness, SMDs, and CVD risk factors remain unknown. Here we explored overlapping genetic architecture and genetic loci shared between SMDs, loneliness, and CVD risk factors. We analyzed large independent genome-wide association study data on schizophrenia (SCZ), bipolar disorder (BD), major depression (MD), loneliness and CVD risk factors using bivariate causal mixture mode (MiXeR), which estimates the total amount of shared variants, and conditional false discovery rate to evaluate overlap in specific loci. We observed substantial genetic overlap between SMDs, loneliness and CVD risk factors, beyond genetic correlation. We identified 149 loci jointly associated with loneliness and SMDs (MD n = 67, SCZ n = 54, and BD n = 28), and 55 distinct loci jointly associated with loneliness and CVD risk factors. A total of 153 novel loneliness loci were found. Most of the shared loci possessed concordant effect directions, suggesting that genetic risk for loneliness may increase the risk of both SMDs and CVD. Functional analyses of the shared loci implicated biological processes related to the brain, metabolic processes, chromatin and immune system. Altogether, the study revealed polygenic overlap between loneliness, SMDs and CVD risk factors, providing new insights into their shared genetic architecture and common genetic mechanisms.
Collapse
|
288
|
Coelho NR, Matos C, Pimpão AB, Correia MJ, Sequeira CO, Morello J, Pereira SA, Monteiro EC. AHR canonical pathway: in vivo findings to support novel antihypertensive strategies. Pharmacol Res 2021; 165:105407. [PMID: 33418029 DOI: 10.1016/j.phrs.2020.105407] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 12/23/2022]
Abstract
Essential hypertension (HTN) is a disease where genetic and environmental factors interact to produce a high prevalent set of almost indistinguishable phenotypes. The weak definition of what is under the umbrella of HTN is a consequence of the lack of knowledge on the players involved in environment-gene interaction and their impact on blood pressure (BP) and mechanisms. The disclosure of these mechanisms that sense and (mal)adapt to toxic-environmental stimuli might at least determine some phenotypes of essential HTN and will have important therapeutic implications. In the present manuscript, we looked closer to the environmental sensor aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor involved in cardiovascular physiology, but better known by its involvement in biotransformation of xenobiotics through its canonical pathway. This review aims to disclose the contribution of the AHR-canonical pathway to HTN. For better mirror the complexity of the mechanisms involved in BP regulation, we privileged evidence from in vivo studies. Here we ascertained the level of available evidence and a comprehensive characterization of the AHR-related phenotype of HTN. We reviewed clinical and rodent studies on AHR-HTN genetic association and on AHR ligands and their impact on BP. We concluded that AHR is a druggable mechanistic linker of environmental exposure to HTN. We conclude that is worth to investigate the canonical pathway of AHR and the expression/polymorphisms of its related genes and/or other biomarkers (e.g. tryptophan-related ligands), in order to identify patients that may benefit from an AHR-centered antihypertensive treatment.
Collapse
Affiliation(s)
- Nuno R Coelho
- Translational Pharmacology Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, Lisboa, 1169-056, Portugal
| | - Clara Matos
- Translational Pharmacology Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, Lisboa, 1169-056, Portugal
| | - António B Pimpão
- Translational Pharmacology Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, Lisboa, 1169-056, Portugal
| | - M João Correia
- Translational Pharmacology Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, Lisboa, 1169-056, Portugal
| | - Catarina O Sequeira
- Translational Pharmacology Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, Lisboa, 1169-056, Portugal
| | - Judit Morello
- Translational Pharmacology Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, Lisboa, 1169-056, Portugal
| | - Sofia A Pereira
- Translational Pharmacology Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, Lisboa, 1169-056, Portugal.
| | - Emília C Monteiro
- Translational Pharmacology Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School
- Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, Lisboa, 1169-056, Portugal
| |
Collapse
|
289
|
Wan EYF, Fung WT, Schooling CM, Au Yeung SL, Kwok MK, Yu EYT, Wang Y, Chan EWY, Wong ICK, Lam CLK. Blood Pressure and Risk of Cardiovascular Disease in UK Biobank: A Mendelian Randomization Study. Hypertension 2021; 77:367-375. [PMID: 33390054 DOI: 10.1161/hypertensionaha.120.16138] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This study aims to evaluate the causal association of blood pressure (BP) with cardiovascular diseases (CVDs). Two-sample Mendelian randomization was performed using a large genome-wide association study (n=299 024) and the UK Biobank cohort (n=375 256). We identified 327 and 364 single-nucleotide polymorphisms strongly and independently associated with systolic BP and diastolic BP, respectively, as genetic instruments to assess the causal association of BP with total CVD, CVD mortality, and 14 cardiovascular conditions. Nonlinearity was examined with nonlinear instrumental variable assumptions. Genetically predicted BP was significantly positively associated with total CVD (systolic BP, per 10 mm Hg: odds ratio [OR], 1.32 [95% CI, 1.25-1.40]; diastolic BP, per 5 mm Hg: OR, 1.20 [95% CI, 1.15-1.26]). Similar positive causal associations were observed for 14 cardiovascular conditions including ischemic heart disease (systolic BP, per 10 mm Hg: OR, 1.33 [95% CI, 1.24-1.41]; diastolic BP, per 5 mm Hg: OR, 1.20 [95% CI, 1.14-1.27]) and stroke (systolic BP, per 10 mm Hg: OR, 1.35 [95% CI, 1.24-1.48]; diastolic BP, per 5 mm Hg: OR, 1.20 [95% CI, 1.12-1.28]). Nonlinearity Mendelian randomization test demonstrated linear causal association of BP with these outcomes. Consistent estimates were observed in sensitivity analyses, suggesting robustness of the associations and minimal horizontal pleiotropy. The linear positive causal association of BP and CVD was consistent with previous findings that lower BP is better, thus consolidating clinical knowledge on hypertension management in CVD risk reduction.
Collapse
Affiliation(s)
- Eric Yuk Fai Wan
- Department of Family Medicine and Primary Care (E.Y.F.W., W.T.F., E.Y.T.Y., Y.W., C.L.K.L.).,Department of Pharmacology and Pharmacy (E.Y.F.W., I.C.K.W.)
| | - Wing Tung Fung
- Department of Family Medicine and Primary Care (E.Y.F.W., W.T.F., E.Y.T.Y., Y.W., C.L.K.L.)
| | - C Mary Schooling
- School of Public Health, Li Ka Shing Faculty of Medicine (C.M.S., S.L.A.Y., M.K.K.).,The University of Hong Kong. School of Public Health and Health Policy, City University of New York (C.M.S.)
| | - Shiu Lun Au Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine (C.M.S., S.L.A.Y., M.K.K.)
| | | | - Esther Yee Tak Yu
- Department of Family Medicine and Primary Care (E.Y.F.W., W.T.F., E.Y.T.Y., Y.W., C.L.K.L.)
| | - Yuan Wang
- Department of Family Medicine and Primary Care (E.Y.F.W., W.T.F., E.Y.T.Y., Y.W., C.L.K.L.)
| | - Esther Wai Yin Chan
- Department of Pharmacology and Pharmacy, Centre for Safe Medication Practice and Research (E.W.Y.C.)
| | - Ian Chi Kei Wong
- Department of Pharmacology and Pharmacy (E.Y.F.W., I.C.K.W.).,Research Department of Practice and Policy, School of Pharmacy, University College London, United Kingdom (I.C.K.W.)
| | - Cindy Lo Kuen Lam
- Department of Family Medicine and Primary Care (E.Y.F.W., W.T.F., E.Y.T.Y., Y.W., C.L.K.L.)
| |
Collapse
|
290
|
Magavern EF, Warren HR, Ng FL, Cabrera CP, Munroe PB, Caulfield MJ. An Academic Clinician's Road Map to Hypertension Genomics: Recent Advances and Future Directions MMXX. Hypertension 2021; 77:284-295. [PMID: 33390048 DOI: 10.1161/hypertensionaha.120.14535] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
At the dawn of the new decade, it is judicious to reflect on the boom of knowledge about polygenic risk for essential hypertension supplied by the wealth of genome-wide association studies. Hypertension continues to account for significant cardiovascular morbidity and mortality, with increasing prevalence anticipated. Here, we overview recent advances in the use of big data to understand polygenic hypertension, as well as opportunities for future innovation to translate this windfall of knowledge into clinical benefit.
Collapse
Affiliation(s)
- Emma F Magavern
- From the William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Helen R Warren
- From the William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Fu L Ng
- From the William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Claudia P Cabrera
- From the William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Patricia B Munroe
- From the William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Mark J Caulfield
- From the William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| |
Collapse
|
291
|
He Y, Zhu H, Zhang M, Li J, Ma S, Lu Y, Chen L, Zhang M, Peng H. Association Between Serum Furin and Fasting Glucose: A Cross-Sectional Study in Chinese Adults. Front Endocrinol (Lausanne) 2021; 12:781890. [PMID: 35046896 PMCID: PMC8761844 DOI: 10.3389/fendo.2021.781890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/08/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Furin has been associated with glucose metabolic phenotypes in small sampled clinical studies. However, this association has not yet been studied in Chinese. Here, we aimed to examine the association between serum furin and fasting glucose in Chinese adults. METHODS Serum furin and fasting plasma glucose were assayed for 2,172 participants (mean aged 53 years, 38% men) in the Gusu cohort. A median regression model was applied to examine the association between serum furin and fasting glucose, adjusting for age, sex, education level, cigarette smoking, alcohol drinking, obesity, blood pressure, and lipids. To facilitate data interpretation, the association between serum furin and prevalent diabetes was also examined. RESULTS Serum furin was negatively associated with fasting glucose (β=-0.18, P<0.001 for log-furin). In participants with diabetes, serum furin was significantly lower than those with normal glucose (median: 0.90 ng/mL vs. 1.05 ng/mL, P=0.001). Compared with participants in the highest quartile of serum furin, those in the lowest quartile had 42% and 80% increased risk of prevalent prediabetes (OR=1.42, 95%CI: 1.05-1.92, P=0.023) and diabetes (OR=1.80, 95%CI: 1.13-2.91, P=0.015), respectively. CONCLUSIONS Serum furin was negatively associated with prediabetes and diabetes in Chinese adults. Our findings suggest that serum furin may be a risk factor or a biomarker of diabetes.
Collapse
Affiliation(s)
- Yan He
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Hanyun Zhu
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Min Zhang
- Central Office, Suzhou National New and Hi-Tech Industrial Development Zone Center for Disease Control and Prevention, Suzhou, China
| | - Jing Li
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Shengqi Ma
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yin Lu
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Linan Chen
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Mingzhi Zhang
- Department of Biostatistics, School of Public Health, Medical College of Soochow University, Suzhou, China
- *Correspondence: Hao Peng, ; Mingzhi Zhang,
| | - Hao Peng
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou, China
- *Correspondence: Hao Peng, ; Mingzhi Zhang,
| |
Collapse
|
292
|
Evidence for a causal association between milk intake and cardiometabolic disease outcomes using a two-sample Mendelian Randomization analysis in up to 1,904,220 individuals. Int J Obes (Lond) 2021; 45:1751-1762. [PMID: 34024907 PMCID: PMC8310799 DOI: 10.1038/s41366-021-00841-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 04/08/2021] [Accepted: 04/26/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND High milk intake has been associated with cardio-metabolic risk. We conducted a Mendelian Randomization (MR) study to obtain evidence for the causal relationship between milk consumption and cardio-metabolic traits using the lactase persistence (LCT-13910 C > T, rs4988235) variant as an instrumental variable. METHODS We tested the association of LCT genotype with milk consumption (for validation) and with cardio-metabolic traits (for a possible causal association) in a meta-analysis of the data from three large-scale population-based studies (1958 British Birth Cohort, Health and Retirement study, and UK Biobank) with up to 417,236 participants and using summary statistics from consortia meta-analyses on intermediate traits (N = 123,665-697,307) and extended to cover disease endpoints (N = 86,995-149,821). RESULTS In the UK Biobank, carriers of 'T' allele of LCT variant were more likely to consume milk (P = 7.02 × 10-14). In meta-analysis including UK Biobank, the 1958BC, the HRS, and consortia-based studies, under an additive model, 'T' allele was associated with higher body mass index (BMI) (Pmeta-analysis = 4.68 × 10-12) and lower total cholesterol (TC) (P = 2.40 × 10-36), low-density lipoprotein cholesterol (LDL-C) (P = 2.08 × 10-26) and high-density lipoprotein cholesterol (HDL-C) (P = 9.40 × 10-13). In consortia meta-analyses, 'T' allele was associated with a lower risk of coronary artery disease (OR:0.86, 95% CI:0.75-0.99) but not with type 2 diabetes (OR:1.06, 95% CI:0.97-1.16). Furthermore, the two-sample MR analysis showed a causal association between genetically instrumented milk intake and higher BMI (P = 3.60 × 10-5) and body fat (total body fat, leg fat, arm fat and trunk fat; P < 1.37 × 10-6) and lower LDL-C (P = 3.60 × 10-6), TC (P = 1.90 × 10-6) and HDL-C (P = 3.00 × 10-5). CONCLUSIONS Our large-scale MR study provides genetic evidence for the association of milk consumption with higher BMI but lower serum cholesterol levels. These data suggest no need to limit milk intakes with respect to cardiovascular disease risk, with the suggested benefits requiring confirmation in further studies.
Collapse
|
293
|
Tang Q, Wang L, Cai R, Zhang L, Zhang X, Liu X, Liu S. The association of MOV10 polymorphism and expression levels with preeclampsia in the Chinese Han population. Mol Genet Genomic Med 2021; 9:e1564. [PMID: 33269545 PMCID: PMC7963431 DOI: 10.1002/mgg3.1564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND To assess the relationship between MOV10 rs2932538 polymorphism and susceptibility to preeclampsia (PE) in the Chinese Han population and to investigate whether the placental expression of MOV10 have association with PE. METHODS We enrolled 1021 pregnant women with PE and 1594 normotensive pregnant women to analyze genotyping of MOV10 rs2932538. Clinical data and related test results of all subjects were collected and analyzed. For volunteers providing placentas, real-time PCR, Western blot, and immunohistochemistry were applied to assess the expression level of MOV10. RESULTS There was significant statistical difference between preeclamptic patients and healthy subjects in genotype distributions and alleles. The frequencies of genotypes TT+CT were significantly associated with the increased risk of preeclampsia. Besides, T alleles were found to be related to a higher risk of PE. Significant statistical difference was also observed on distributions of genotype in PE without/with severe features group compared or early onset/late onset versus controls. The placental expression of MOV10 was lower in preeclamptic women, however, no relationship was found between MOV10 expression level and MOV10 rs2932538 genotypes. CONCLUSION This study suggests that MOV10 rs2932538 polymorphism may be associated with PE susceptibility in the Chinese Han population. The placental expression of MOV10 decrease in PE but have no relationship with rs2932538 polymorphism.
Collapse
Affiliation(s)
- Qian Tang
- Medical Genetic Departmentthe Affiliated Hospital of Qingdao UniversityQingdaoChina
- Prenatal Diagnosis Centerthe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Ling Wang
- Department of Nephrologythe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Renmei Cai
- Prenatal Diagnosis CenterQingdao Municipal HospitalQingdaoChina
| | - Lu Zhang
- Medical Genetic Departmentthe Affiliated Hospital of Qingdao UniversityQingdaoChina
- Prenatal Diagnosis Centerthe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Xiaoxiao Zhang
- Department of Nephrologythe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Xuemei Liu
- Department of Nephrologythe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Shiguo Liu
- Medical Genetic Departmentthe Affiliated Hospital of Qingdao UniversityQingdaoChina
- Prenatal Diagnosis Centerthe Affiliated Hospital of Qingdao UniversityQingdaoChina
| |
Collapse
|
294
|
S Machado IB, Tofanelli MR, Saldanha da Silva AA, Simões E Silva AC. Factors Associated with Primary Hypertension in Pediatric Patients: An Up-to-Date. Curr Pediatr Rev 2021; 17:15-37. [PMID: 33430749 DOI: 10.2174/1573396317999210111200222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Arterial hypertension in children is considered a common alteration nowadays, mainly because obesity is a growing worldwide problem closely related to increased blood pressure. Childhood hypertension can be classified as primary or secondary, depending on the etiology. Primary or essential hypertension still has its pathophysiology not fully elucidated, and there is no consensus in the literature on most underlying mechanisms. In this review, genetic and environmental factors, including sodium and potassium intake, socioeconomic status, ethnicity, family structure, obesity, sedentary lifestyle, prematurity and low birth weight, prenatal and postnatal exposures are highlighted. OBJECTIVE The present study aimed to perform an update on primary hypertension in childhood, providing clinicians and researchers an overview of the current state of the literature regarding the influence of genetic and environmental factors. METHODS This integrative review searched for articles on genetic and environmental factors related to primary hypertension in pediatric patients. The databases evaluated were PubMed and Scopus. RESULTS The studies have provided insights regarding many genetic and environmental factors, in addition to their association with the pathophysiology of primary hypertension in childhood. Findings corroborated the idea that primary hypertension is a multifactorial disease. Further studies in the pediatric population are needed to elucidate the underlying mechanisms. CONCLUSION The study of primary hypertension in pediatrics has utmost importance for the adoption of preventive measures and the development of more efficient treatments, therefore reducing childhood morbidity and the incidence of cardiovascular diseases and other health consequences later in life.
Collapse
Affiliation(s)
- Isabella Barreto S Machado
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Matheus Rampinelli Tofanelli
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ariadna A Saldanha da Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
295
|
Ahisar Y, Thanassoulis G, Huang KN, Ohayon SM, Afilalo J. Intersecting Genetics of Frailty and Cardiovascular Disease. J Nutr Health Aging 2021; 25:1023-1027. [PMID: 34545923 DOI: 10.1007/s12603-021-1673-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To determine the genetic correlates of physical frailty and sarcopenia, focusing on single nucleotide polymorphisms (SNPs) in genome-wide association studies (GWAS), and to explore the genetic overlap of frailty with cardiovascular disease (CVD) and its risk factors. METHODS PubMed was systematically searched for GWAS studies investigating the association between SNPs and objective measures of physical frailty or sarcopenia. SNPs were retained if they were associated with one of the phenotypes of interest by a p-value of 5.0x10-8 or less. RESULTS Ten studies were included, with a total of 237 SNPs in 181 genes being associated with physical frailty or sarcopenia; as measured by handgrip strength or lean (muscle) mass. These genes were cross-referenced in the GWAS Catalog, and many of them were found to be associated with CVD or metabolic syndrome. CONCLUSIONS Evidence from GWAS has shown that frailty is associated with common genetic polymorphisms. Many of these polymorphisms have been implicated in CVD, supporting the hypothesis of a shared pathophysiology between these entities. Future studies are eagerly anticipated to map out the mechanistic links and discover therapeutic targets and novel biomarkers for frailty.
Collapse
Affiliation(s)
- Y Ahisar
- Jonathan Afilalo, MD, MSc, FACC, FRCPC, Associate Professor, McGill University, Director, Geriatric Cardiology Fellowship Program, Jewish General Hospital, 3755 Cote Ste Catherine Rd, E-222, Montreal, QC H3T 1E2, Phone: (514) 340-7540 | Fax: (514) 340-7534,
| | | | | | | | | |
Collapse
|
296
|
Choi I, Beedholm K, Dam VS, Bae SH, Noble DJ, Garraway SM, Aalkjaer C, Boedtkjer E. Sodium bicarbonate cotransporter NBCn1/Slc4a7 affects locomotor activity and hearing in mice. Behav Brain Res 2020; 401:113065. [PMID: 33321164 PMCID: PMC9069564 DOI: 10.1016/j.bbr.2020.113065] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 11/19/2020] [Accepted: 12/08/2020] [Indexed: 02/06/2023]
Abstract
Despite a widespread expression pattern in the central nervous system, the role of the sodium bicarbonate cotransporter NBCn1/Slc4a7 has not been investigated for locomotor activity, emotion and cognition. Here, we addressed the behavioral consequences of NBCn1 knockout and evaluated hearing and vision that are reportedly impaired in an earlier line of NBCn1 knockout mice and may contribute to behavioral changes. In a circular open field, the knockout mice traveled a shorter distance, especially in the periphery of the chamber, than wildtype littermates. The knockout mice also traveled a shorter total distance in a home cage-like open field. Rearing and grooming behaviors were reduced. The knockout and control mice displayed similar time spent and number of open and closed arms in the elevated plus maze test, indicating negligible change in anxiety. In the Morris water maze test, both groups of mice learned the location of an escape platform within comparable time on the training trials and showed similar platform identification on the probe trial. The knockout mice maintained normal visual responses in the optokinetic drum and produced evoked potentials in response to light stimuli. However, these mice failed to produce auditory evoked potentials. qPCR revealed a robust expression of an alternatively transcribed NBCn1 variant in the knockout mouse retina. These results indicate that NBCn1 deletion leads to reduced locomotor activity in mice by affecting their exploratory behaviors or emotionality. The deletion also causes hearing loss, but its effect on vision varies between different lines of knockout mice.
Collapse
Affiliation(s)
- Inyeong Choi
- Department of Physiology, Emory University School of Medicine, Atlanta, USA.
| | | | - Vibeke S Dam
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Seong-Ho Bae
- Department of Medicine, Division of Cardiology, Emory University School of Medicine, Atlanta, USA
| | - Donald J Noble
- Department of Physiology, Emory University School of Medicine, Atlanta, USA
| | - Sandra M Garraway
- Department of Physiology, Emory University School of Medicine, Atlanta, USA
| | | | - Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
297
|
Bykhovskaya Y, Rabinowitz YS. Update on the genetics of keratoconus. Exp Eye Res 2020; 202:108398. [PMID: 33316263 DOI: 10.1016/j.exer.2020.108398] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
In the past few years we have seen a great acceleration of discoveries in the field of keratoconus including new treatments, diagnostic tools, genomic and molecular determinants of disease risk. Recent genome-wide association studies (GWAS) of keratoconus cases and population wide studies of variation in central corneal thickness and in corneal biomechanical properties confirmed already identified genes and found many new susceptibility variants and biological pathways. Recent findings in genetic determinants of familial keratoconus revealed functionally important variants and established first mouse model of keratoconus. Latest transcriptomic and expression studies started assessing novel non-coding RNA targets in addition to identifying tissue specific effects of coding genes. First genomic insights into better prediction of treatment outcomes are bringing the advent of genomic medicine into keratoconus clinical practice.
Collapse
Affiliation(s)
- Yelena Bykhovskaya
- Cornea Genetic Eye Institute, Department of Surgery and Board of the Governors Regenerative Medicine Institute, Beverly Hills, Cedars-Sinai, Los Angeles, CA, United States.
| | - Yaron S Rabinowitz
- Cornea Genetic Eye Institute, Department of Surgery and Board of the Governors Regenerative Medicine Institute, Beverly Hills, Cedars-Sinai, Los Angeles, CA, United States
| |
Collapse
|
298
|
Sargurupremraj M, Suzuki H, Jian X, Sarnowski C, Evans TE, Bis JC, Eiriksdottir G, Sakaue S, Terzikhan N, Habes M, Zhao W, Armstrong NJ, Hofer E, Yanek LR, Hagenaars SP, Kumar RB, van den Akker EB, McWhirter RE, Trompet S, Mishra A, Saba Y, Satizabal CL, Beaudet G, Petit L, Tsuchida A, Zago L, Schilling S, Sigurdsson S, Gottesman RF, Lewis CE, Aggarwal NT, Lopez OL, Smith JA, Valdés Hernández MC, van der Grond J, Wright MJ, Knol MJ, Dörr M, Thomson RJ, Bordes C, Le Grand Q, Duperron MG, Smith AV, Knopman DS, Schreiner PJ, Evans DA, Rotter JI, Beiser AS, Maniega SM, Beekman M, Trollor J, Stott DJ, Vernooij MW, Wittfeld K, Niessen WJ, Soumaré A, Boerwinkle E, Sidney S, Turner ST, Davies G, Thalamuthu A, Völker U, van Buchem MA, Bryan RN, Dupuis J, Bastin ME, Ames D, Teumer A, Amouyel P, Kwok JB, Bülow R, Deary IJ, Schofield PR, Brodaty H, Jiang J, Tabara Y, Setoh K, Miyamoto S, Yoshida K, Nagata M, Kamatani Y, Matsuda F, Psaty BM, Bennett DA, De Jager PL, Mosley TH, Sachdev PS, Schmidt R, Warren HR, Evangelou E, Trégouët DA, Ikram MA, Wen W, DeCarli C, Srikanth VK, Jukema JW, Slagboom EP, Kardia SLR, Okada Y, Mazoyer B, et alSargurupremraj M, Suzuki H, Jian X, Sarnowski C, Evans TE, Bis JC, Eiriksdottir G, Sakaue S, Terzikhan N, Habes M, Zhao W, Armstrong NJ, Hofer E, Yanek LR, Hagenaars SP, Kumar RB, van den Akker EB, McWhirter RE, Trompet S, Mishra A, Saba Y, Satizabal CL, Beaudet G, Petit L, Tsuchida A, Zago L, Schilling S, Sigurdsson S, Gottesman RF, Lewis CE, Aggarwal NT, Lopez OL, Smith JA, Valdés Hernández MC, van der Grond J, Wright MJ, Knol MJ, Dörr M, Thomson RJ, Bordes C, Le Grand Q, Duperron MG, Smith AV, Knopman DS, Schreiner PJ, Evans DA, Rotter JI, Beiser AS, Maniega SM, Beekman M, Trollor J, Stott DJ, Vernooij MW, Wittfeld K, Niessen WJ, Soumaré A, Boerwinkle E, Sidney S, Turner ST, Davies G, Thalamuthu A, Völker U, van Buchem MA, Bryan RN, Dupuis J, Bastin ME, Ames D, Teumer A, Amouyel P, Kwok JB, Bülow R, Deary IJ, Schofield PR, Brodaty H, Jiang J, Tabara Y, Setoh K, Miyamoto S, Yoshida K, Nagata M, Kamatani Y, Matsuda F, Psaty BM, Bennett DA, De Jager PL, Mosley TH, Sachdev PS, Schmidt R, Warren HR, Evangelou E, Trégouët DA, Ikram MA, Wen W, DeCarli C, Srikanth VK, Jukema JW, Slagboom EP, Kardia SLR, Okada Y, Mazoyer B, Wardlaw JM, Nyquist PA, Mather KA, Grabe HJ, Schmidt H, Van Duijn CM, Gudnason V, Longstreth WT, Launer LJ, Lathrop M, Seshadri S, Tzourio C, Adams HH, Matthews PM, Fornage M, Debette S. Cerebral small vessel disease genomics and its implications across the lifespan. Nat Commun 2020; 11:6285. [PMID: 33293549 PMCID: PMC7722866 DOI: 10.1038/s41467-020-19111-2] [Show More Authors] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 09/10/2020] [Indexed: 12/14/2022] Open
Abstract
White matter hyperintensities (WMH) are the most common brain-imaging feature of cerebral small vessel disease (SVD), hypertension being the main known risk factor. Here, we identify 27 genome-wide loci for WMH-volume in a cohort of 50,970 older individuals, accounting for modification/confounding by hypertension. Aggregated WMH risk variants were associated with altered white matter integrity (p = 2.5×10-7) in brain images from 1,738 young healthy adults, providing insight into the lifetime impact of SVD genetic risk. Mendelian randomization suggested causal association of increasing WMH-volume with stroke, Alzheimer-type dementia, and of increasing blood pressure (BP) with larger WMH-volume, notably also in persons without clinical hypertension. Transcriptome-wide colocalization analyses showed association of WMH-volume with expression of 39 genes, of which four encode known drug targets. Finally, we provide insight into BP-independent biological pathways underlying SVD and suggest potential for genetic stratification of high-risk individuals and for genetically-informed prioritization of drug targets for prevention trials.
Collapse
Affiliation(s)
- Muralidharan Sargurupremraj
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
| | - Hideaki Suzuki
- Tohoku Medical Megabank Organization, Tohoku University, 2-1, Seiryo, Aoba, Sendai, 980-8573, Japan
- Department of Cardiovascular Medicine, Tohoku University Hospital, 1-1, Seiryo, Aoba, Sendai, 980-8574, Japan
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK
| | - Xueqiu Jian
- University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, 77030, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, 78229, USA
| | - Chloé Sarnowski
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Tavia E Evans
- Department of Clinical Genetics, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, 98101, USA
| | | | - Saori Sakaue
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
- Department of Allergy and Rheumatology, Graduate School of Medicine, the University of Tokyo, Tokyo, 113-0033, Japan
| | - Natalie Terzikhan
- Department of Epidemiology, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Mohamad Habes
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, 78229, USA
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Institute for Community Medicine, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109-2029, USA
| | - Nicola J Armstrong
- Mathematics and Statistics, Murdoch University, Murdoch, WA, 6150, Australia
| | - Edith Hofer
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University of Graz, 8036, Graz, Austria
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, 8036, Graz, Austria
| | - Lisa R Yanek
- GeneSTAR Research Program, Division of General Internal Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Saskia P Hagenaars
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Social Genetic and Developmental Psychiatry Research Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 8AF, UK
| | - Rajan B Kumar
- Department of Public Health Sciences, University of California at Davis, Davis, CA, 95616, USA
| | - Erik B van den Akker
- Section of Molecular Epidemiology, Biomedical Sciences, Leiden university Medical Center, 2333 ZA, Leiden, The Netherlands
- Pattern Recognition & Bioinformatics, Delft University of Technology, Delft, NL, 2629 HS, USA
- Leiden Computational Biology Centre, Leiden University Medical Centre, 2333 ZA, Leiden, The Netherlands
| | - Rebekah E McWhirter
- Centre for Law and Genetics, Faculty of Law, University of Tasmania, Hobart, TAS, 7005, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Stella Trompet
- Department of Internal Medicine, section of gerontology and geriatrics, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
- Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Aniket Mishra
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
| | - Yasaman Saba
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
- Gottfried Schatz Research Center, Department of Molecular Biology and Biochemistry, Medical University of Graz, 8010, Graz, Austria
| | - Claudia L Satizabal
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, 78229, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, 02215, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Gregory Beaudet
- University of Bordeaux, IMN, UMR 5293, 33000, Bordeaux, France
| | - Laurent Petit
- University of Bordeaux, IMN, UMR 5293, 33000, Bordeaux, France
| | - Ami Tsuchida
- University of Bordeaux, IMN, UMR 5293, 33000, Bordeaux, France
| | - Laure Zago
- University of Bordeaux, IMN, UMR 5293, 33000, Bordeaux, France
| | - Sabrina Schilling
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
| | | | | | - Cora E Lewis
- University of Alabama at Birmingham School of Medicine, Birmingham, AL, 35233, USA
| | - Neelum T Aggarwal
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Oscar L Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109-2029, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - Maria C Valdés Hernández
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Row Fogo Centre for Ageing and The Brain, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Jeroen van der Grond
- Department of Radiology, Leiden University medical Center, 2333 ZA, Leiden, The Netherlands
| | - Margaret J Wright
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
- Centre for Advanced Imaging, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Maria J Knol
- Department of Epidemiology, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Marcus Dörr
- Department of Internal Medicine B, University Medicine Greifswald, 17475, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), partner site Greifswald, 17475, Greifswald, Germany
| | - Russell J Thomson
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
- Centre for Research in Mathematics and Data Science, Western Sydney University, Penrith, NSW, 2751, Australia
| | - Constance Bordes
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
| | - Quentin Le Grand
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
| | - Marie-Gabrielle Duperron
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
| | | | | | - Pamela J Schreiner
- University of Minnesota School of Public Health, Minneapolis, MN, 55455, USA
| | - Denis A Evans
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Alexa S Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, 02215, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Susana Muñoz Maniega
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Marian Beekman
- Section of Molecular Epidemiology, Biomedical Sciences, Leiden university Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Julian Trollor
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, 2052, Australia
- Department of Developmental Disability Neuropsychiatry, School of Psychiatry, University of New South Wales, Sydney, NSW, 2052, Australia
| | - David J Stott
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Meike W Vernooij
- Department of Radiology & Nuclear Medicine, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Katharina Wittfeld
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, 17489, Greifswald, Germany
| | - Wiro J Niessen
- Department of Radiology & Nuclear Medicine, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
- Faculty of Applied Sciences, Delft University of Technology, Delft, NL, 2629 HS, USA
| | - Aicha Soumaré
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
| | - Eric Boerwinkle
- University of Texas Health Science Center at Houston School of Public Health, Houston, TX, 77030, USA
| | - Stephen Sidney
- Kaiser Permanente Division of Research, Oakland, CA, 94612, USA
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, 55905, USA
| | - Gail Davies
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, 8036, Graz, Austria
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Anbupalam Thalamuthu
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Mark A van Buchem
- Row Fogo Centre for Ageing and The Brain, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - R Nick Bryan
- The University of Texas at Austin Dell Medical School, Austin, TX, 78712, USA
| | - Josée Dupuis
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, 78229, USA
- Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Mark E Bastin
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, 8036, Graz, Austria
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, 48104, USA
| | - David Ames
- National Ageing Research Institute Royal Melbourne Hospital, Parkville, VIC, 3052, Australia
- Academic Unit for Psychiatry of Old Age, University of Melbourne, St George's Hospital, Kew, VIC, 3101, Australia
| | - Alexander Teumer
- Department of Epidemiology, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
- Department of Internal Medicine B, University Medicine Greifswald, 17475, Greifswald, Germany
| | - Philippe Amouyel
- Inserm U1167, 59000, Lille, France
- Department of Epidemiology and Public Health, Pasteur Institute of Lille, 59000, Lille, France
| | - John B Kwok
- Brain and Mind Centre - The University of Sydney, Camperdown, NSW, 2050, Australia
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Robin Bülow
- Department of Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, 17489, Greifswald, Germany
| | - Ian J Deary
- Institute for Medical Informatics, Statistics and Documentation, Medical University of Graz, 8036, Graz, Austria
- Department of Psychology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
| | - Peter R Schofield
- School of Medical Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
- Neuroscience Research Australia, Randwick, NSW, 2031, Australia
| | - Henry Brodaty
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
- Dementia Centre for Research Collaboration, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jiyang Jiang
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
| | - Yasuharu Tabara
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Kazuya Setoh
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Susumu Miyamoto
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Kazumichi Yoshida
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Manabu Nagata
- Department of Neurosurgery, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Yoichiro Kamatani
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
| | - Fumihiko Matsuda
- Center for Genomic Medicine, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Bruce M Psaty
- Departments of Epidemiology, Medicine and Health Services, University of Washington, Seattle, WA, 98195, USA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA, 98101, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Medical Center, New York, NY, 10032, USA
- Program in Population and Medical Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Thomas H Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Perminder S Sachdev
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute and Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, NSW, 2031, Australia
| | - Reinhold Schmidt
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109-2029, USA
| | - Helen R Warren
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, E1 4NS, UK
- National Institute for Health Research Barts Cardiovascular Biomedical Research Unit, Queen Mary University of London, London, EC1M 6BQ, UK
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Mpizani, 455 00, Greece
| | - David-Alexandre Trégouët
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
| | - Mohammad A Ikram
- Department of Epidemiology, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Wei Wen
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Charles DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Sacramento, CA, 95817, USA
| | - Velandai K Srikanth
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, 7000, Australia
- Peninsula Clinical School, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - J Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Eline P Slagboom
- Section of Molecular Epidemiology, Biomedical Sciences, Leiden university Medical Center, 2333 ZA, Leiden, The Netherlands
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, 48109-2029, USA
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, 565-0871, Japan
- Laboratory for Statistical Analysis, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama City, Kanagawa, 230-0045, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, 565-0871, Osaka, Japan
| | - Bernard Mazoyer
- University of Bordeaux, IMN, UMR 5293, 33000, Bordeaux, France
| | - Joanna M Wardlaw
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
- Row Fogo Centre for Ageing and The Brain, University of Edinburgh, Edinburgh, EH8 9JZ, UK
- MRC UK Dementia Research Institute at the University of Edinburgh, Edinburgh, EH8 9YL, UK
| | - Paul A Nyquist
- Department of Neurology, Johns Hopkins School of Medicine, Baltimone, MD, 21205, USA
- General Internal Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW, 2052, Australia
- Neuroscience Research Australia, Randwick, NSW, 2031, Australia
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475, Greifswald, Germany
- German Center for Neurodegenerative Diseases (DZNE), Rostock/Greifswald, 17475, Greifswald, Germany
| | - Helena Schmidt
- Gottfried Schatz Research Center, Department of Molecular Biology and Biochemistry, Medical University of Graz, 8010, Graz, Austria
| | - Cornelia M Van Duijn
- Nuffield Department of Population Health, University of Oxford, Oxford, OX3 7LF, UK
| | - Vilmundur Gudnason
- Icelandic Heart Association, IS-201, Kópavogur, Iceland
- University of Iceland, Faculty of Medicine, 101, Reykjavík, Iceland
| | - William T Longstreth
- Departments of Neurology and Epidemiology, University of Washington, Seattle, WA, 98104-2420, USA
| | - Lenore J Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute of Aging, The National Institutes of Health, Bethesda, MD, 20892, USA
- Intramural Research Program/National Institute on Aging/National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mark Lathrop
- University of McGill Genome Center, Montreal, QC, H3A 0G1, Canada
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, 78229, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, 02215, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Christophe Tzourio
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France
- CHU de Bordeaux, Pole de santé publique, Service d'information médicale, 33000, Bordeaux, France
| | - Hieab H Adams
- Department of Clinical Genetics, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
- Department of Radiology & Nuclear Medicine, Erasmus MC, 3015 GE, Rotterdam, The Netherlands
| | - Paul M Matthews
- Department of Brain Sciences, Imperial College London, London, W12 0NN, UK
- UK Dementia Research Institute, London, WC1E 6BT, UK
- Data Science Institute, Imperial College London, London, SW7 2AZ, UK
| | - Myriam Fornage
- University of Texas Health Science Center at Houston McGovern Medical School, Houston, TX, 77030, USA.
| | - Stéphanie Debette
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, 33000, Bordeaux, France.
- Department of Neurology, Boston University School of Medicine, Boston, MA, 02118, USA.
- Department of Neurology, CHU de Bordeaux, 33000, Bordeaux, France.
| |
Collapse
|
299
|
Caylak G, Tastan O, Cicek AE. Potpourri: An Epistasis Test Prioritization Algorithm via Diverse SNP Selection. J Comput Biol 2020; 28:365-377. [PMID: 33275856 DOI: 10.1089/cmb.2020.0429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Genome-wide association studies (GWAS) explain a fraction of the underlying heritability of genetic diseases. Investigating epistatic interactions between two or more loci help to close this gap. Unfortunately, the sheer number of loci combinations to process and hypotheses prohibit the process both computationally and statistically. Epistasis test prioritization algorithms rank likely epistatic single nucleotide polymorphism (SNP) pairs to limit the number of tests. However, they still suffer from very low precision. It was shown in the literature that selecting SNPs that are individually correlated with the phenotype and also diverse with respect to genomic location leads to better phenotype prediction due to genetic complementation. Here, we propose that an algorithm that pairs SNPs from such diverse regions and ranks them can improve prediction power. We propose an epistasis test prioritization algorithm that optimizes a submodular set function to select a diverse and complementary set of genomic regions that span the underlying genome. The SNP pairs from these regions are then further ranked w.r.t. their co-coverage of the case cohort. We compare our algorithm with the state of the art on three GWAS and show that (1) we substantially improve precision (from 0.003 to 0.652) while maintaining the significance of selected pairs, (2) decrease the number of tests by 25-fold, and (3) decrease the runtime by 4-fold. We also show that promoting SNPs from regulatory/coding regions improves the performance (up to 0.8). Potpourri is available at http:/ciceklab.cs.bilkent.edu.tr/potpourri.
Collapse
Affiliation(s)
- Gizem Caylak
- Computer Engineering Department, Bilkent University, Ankara, Turkey
| | - Oznur Tastan
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey
| | - A Ercument Cicek
- Computer Engineering Department, Bilkent University, Ankara, Turkey
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
300
|
Chen H, Wang T, Yang J, Huang S, Zeng P. Improved Detection of Potentially Pleiotropic Genes in Coronary Artery Disease and Chronic Kidney Disease Using GWAS Summary Statistics. Front Genet 2020; 11:592461. [PMID: 33343632 PMCID: PMC7744760 DOI: 10.3389/fgene.2020.592461] [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: 08/07/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
The coexistence of coronary artery disease (CAD) and chronic kidney disease (CKD) implies overlapped genetic foundation. However, the common genetic determination between the two diseases remains largely unknown. Relying on summary statistics publicly available from large scale genome-wide association studies (n = 184,305 for CAD and n = 567,460 for CKD), we observed significant positive genetic correlation between CAD and CKD (rg = 0.173, p = 0.024) via the linkage disequilibrium score regression. Next, we implemented gene-based association analysis for each disease through MAGMA (Multi-marker Analysis of GenoMic Annotation) and detected 763 and 827 genes associated with CAD or CKD (FDR < 0.05). Among those 72 genes were shared between the two diseases. Furthermore, by integrating the overlapped genetic information between CAD and CKD, we implemented two pleiotropy-informed informatics approaches including cFDR (conditional false discovery rate) and GPA (Genetic analysis incorporating Pleiotropy and Annotation), and identified 169 and 504 shared genes (FDR < 0.05), of which 121 genes were simultaneously discovered by cFDR and GPA. Importantly, we found 11 potentially new pleiotropic genes related to both CAD and CKD (i.e., ARHGEF19, RSG1, NDST2, CAMK2G, VCL, LRP10, RBM23, USP10, WNT9B, GOSR2, and RPRML). Five of the newly identified pleiotropic genes were further repeated via an additional dataset CAD available from UK Biobank. Our functional enrichment analysis showed that those pleiotropic genes were enriched in diverse relevant pathway processes including quaternary ammonium group transmembrane transporter, dopamine transport. Overall, this study identifies common genetic architectures overlapped between CAD and CKD and will help to advance understanding of the molecular mechanisms underlying the comorbidity of the two diseases.
Collapse
Affiliation(s)
- Haimiao Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Ting Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Jinna Yang
- Department of Infectious Diseases, People's Hospital of Zhuji, Shaoxing, China
| | - Shuiping Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China.,Center for Medical Statistics and Data Analysis, School of Public Health, Xuzhou Medical University, Xuzhou, China
| | - Ping Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, China.,Center for Medical Statistics and Data Analysis, School of Public Health, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|