1
|
Clavell-Revelles P, Reese F, Carbonell-Sala S, Degalez F, Oliveros W, Arnan C, Guigó R, Melé M. Long-read transcriptomics of a diverse human cohort reveals widespread ancestry bias in gene annotation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643250. [PMID: 40166264 PMCID: PMC11956941 DOI: 10.1101/2025.03.14.643250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Accurate gene annotations are fundamental for interpreting genetic variation, cellular function, and disease mechanisms. However, current human gene annotations are largely derived from transcriptomic data of individuals with European ancestry, introducing potential biases that remain uncharacterized. Here, we generate over 800 million full-length reads with long-read RNA-seq in 43 lymphoblastoid cell line samples from eight genetically-diverse human populations and build a cross-ancestry gene annotation. We show that transcripts from non-European samples are underrepresented in reference gene annotations, leading to systematic biases in allele-specific transcript usage analyses. Furthermore, we show that personal genome assemblies enhance transcript discovery compared to the generic GRCh38 reference assembly, even though genomic regions unique to each individual are heavily depleted of genes. These findings underscore the urgent need for a more inclusive gene annotation framework that accurately represents global transcriptome diversity.
Collapse
Affiliation(s)
- Pau Clavell-Revelles
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Catalonia
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
- Universitat de Barcelona (UB), Barcelona, Catalonia
| | - Fairlie Reese
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Catalonia
| | - Sílvia Carbonell-Sala
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
| | - Fabien Degalez
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
| | - Winona Oliveros
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Catalonia
- Universitat de Barcelona (UB), Barcelona, Catalonia
| | - Carme Arnan
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
- Universitat Pompeu Fabra (UPF), Barcelona, Catalonia
| | - Marta Melé
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Catalonia
| |
Collapse
|
2
|
Little A, Zhao N, Mikhaylova A, Zhang A, Ling W, Thibord F, Johnson AD, Raffield LM, Curran JE, Blangero J, O'Connell JR, Xu H, Rotter JI, Rich SS, Rice KM, Chen MH, Reiner A, Kooperberg C, Vu T, Hou L, Fornage M, Loos RJF, Kenny E, Mathias R, Becker L, Smith AV, Boerwinkle E, Yu B, Thornton T, Wu MC. General Kernel Machine Methods for Multi-Omics Integration and Genome-Wide Association Testing With Related Individuals. Genet Epidemiol 2025; 49:e22610. [PMID: 39812506 DOI: 10.1002/gepi.22610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 09/18/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Integrating multi-omics data may help researchers understand the genetic underpinnings of complex traits and diseases. However, the best ways to integrate multi-omics data and use them to address pressing scientific questions remain a challenge. One important and topical problem is how to assess the aggregate effect of multiple genomic data types (e.g. genotypes and gene expression levels) on a phenotype, particularly while accommodating routine issues, such as having related subjects' data in analyses. In this paper, we extend an existing composite kernel machine regression model to integrate two multi-omics data types, while accommodating for general correlation structures amongst outcomes. Due to the kernel machine regression framework, our methods allow for the integration of high-dimensional omics data with small, nonlinear, and interactive effects, and accommodation of general study designs. Here, we focus on scientific questions that aim to assess the association between a functional grouping (such as a gene or a pathway) and a quantitative trait of interest. We use a kernel machine regression to integrate the two multi-omics data types, as they may relate to the trait, and perform a global test of association. We demonstrate the advantage of this approach over single data type association tests via simulation. Finally, we apply this method to a large, multi-ethnic data set to investigate how predicted gene expression and rare genetic variation may be related to two platelet traits.
Collapse
Grants
- U.S. Department of Health and Human Services, National Institute on Minority Health and Health Disparities, National Institutes of Health, National Human Genome Research Institute, National Center for Research Resources, COPD Foundation, National Heart, Lung, and Blood Institute, National Science Foundation, National Institute on Aging, and National Institute of Neurological Disorders and Stroke.
Collapse
Affiliation(s)
- Amarise Little
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Ni Zhao
- Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Anna Mikhaylova
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Angela Zhang
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Wodan Ling
- Department of Population Health Sciences, Division of Biostatistics, Weill Cornell Medicine, New York, New York, USA
| | - Florian Thibord
- National Heart, Lung, and Blood Institute, Boston University's Framingham Heart Study, Framingham, Massachusetts, USA
- National Heart, Lung and Blood Institute, Population Sciences Branch, Framingham, Massachusetts, USA
| | - Andrew D Johnson
- National Heart, Lung, and Blood Institute, Boston University's Framingham Heart Study, Framingham, Massachusetts, USA
- National Heart, Lung and Blood Institute, Population Sciences Branch, Framingham, Massachusetts, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joanne E Curran
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, USA
| | - John Blangero
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, Texas, USA
| | | | - Huichun Xu
- Department of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, Virginia, USA
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Ming-Huei Chen
- National Heart, Lung, and Blood Institute, Boston University's Framingham Heart Study, Framingham, Massachusetts, USA
- National Heart, Lung and Blood Institute, Population Sciences Branch, Framingham, Massachusetts, USA
| | - Alexander Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Thao Vu
- Department of Biostatistics and Informatics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Myriam Fornage
- Brown Foundation Institute for Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Eimear Kenny
- The Charles Bronfman Institute of Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Center for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Pamela Sklar Division of Psychiatric Genomics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rasika Mathias
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lewis Becker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Albert V Smith
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, Michigan, USA
| | - Eric Boerwinkle
- Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Bing Yu
- Department of Epidemiology, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Timothy Thornton
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Michael C Wu
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| |
Collapse
|
3
|
Shapiro MD, Haddad TM, Weintraub HS, Baum SJ, Abdul-Nour K, Sarwat S, Paluy V, Boatwright W, Browne A, Ayaz I, Abbas CA, Ballantyne CM. Lipoprotein(a) levels in a population with clinical atherosclerotic cardiovascular disease in the United States: A subanalysis from the Lp(a)HERITAGE study. J Clin Lipidol 2025; 19:28-38. [PMID: 39909772 DOI: 10.1016/j.jacl.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 02/07/2025]
Abstract
BACKGROUND Elevated lipoprotein(a) (Lp[a]) is the most common inherited dyslipidemia that is independently and causally associated with increased atherosclerotic cardiovascular disease (ASCVD) risk. However, data from diverse populations with ASCVD are lacking. OBJECTIVE To evaluate Lp(a) levels in a diverse, contemporary United States (US) population with ASCVD, stratified by race, ethnicity, and sex. METHODS Lp(a)HERITAGE (NCT03887520) was a multicenter study that estimated the prevalence of elevated Lp(a) in adults (18-80 years) with ASCVD. US participants with Lp(a) measured in nmol/L pre- or post-enrollment were included in this subanalysis. This study was descriptive; therefore, no statistical comparisons were made. RESULTS Of all US participants, 14% had an Lp(a) measurement pre-enrollment. This subanalysis included 7679 US participants with Lp(a) measurements in nmol/L (80.5% White; 66.4% male; mean age 63.8 years [standard deviation ± 9.7]). Median Lp(a) was > 2.5-fold higher in Black participants (132.0 nmol/L; IQR, 57.1-239.6) vs the overall population (52.1 nmol/L; IQR, 15.7-167.8), and higher in females compared with males (69.4 nmol/L; IQR, 20.1-194.7 vs 45.6 nmol/L; IQR, 14.0-152.6, respectively). Lp(a) levels ≥ 125 nmol/L were more prevalent among Black (52.0%) and female (38.9%) participants vs the overall population (33.3%). CONCLUSION In US Lp(a)HERITAGE participants, only 14% had an Lp(a) measurement pre-enrollment, despite having ASCVD. One-third of participants demonstrated Lp(a) levels ≥ 125 nmol/L, the threshold for high ASCVD risk, which was higher among Black (1/2) and female (2/5) participants, suggesting a greater need for Lp(a) testing in these groups to inform ASCVD risk mitigation.
Collapse
Affiliation(s)
- Michael D Shapiro
- Section of Cardiovascular Medicine, Center for Prevention of Cardiovascular Disease, Wake Forest University School of Medicine, Winston-Salem, NC, USA (Dr Shapiro).
| | - Tariq M Haddad
- Virginia Heart, Falls Church, VA, USA (Dr Haddad); Inova Schar Heart and Vascular, Falls Church, VA, USA (Dr Haddad)
| | - Howard S Weintraub
- NYU Langone Center for the Prevention of Cardiovascular Disease, New York, NY, USA (Dr Weintraub)
| | - Seth J Baum
- Flourish Research, Boca Raton, FL, USA (Dr Baum)
| | - Khaled Abdul-Nour
- Division of Cardiovascular Medicine, Henry Ford Hospital, Detroit, MI, USA (Dr Abdul-Nour)
| | - Samiha Sarwat
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA (Drs Sarwat, Paluy, Boatwright, Browne, Ayaz, Abbas)
| | - Vadim Paluy
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA (Drs Sarwat, Paluy, Boatwright, Browne, Ayaz, Abbas)
| | - Wess Boatwright
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA (Drs Sarwat, Paluy, Boatwright, Browne, Ayaz, Abbas)
| | - Auris Browne
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA (Drs Sarwat, Paluy, Boatwright, Browne, Ayaz, Abbas)
| | - Imran Ayaz
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA (Drs Sarwat, Paluy, Boatwright, Browne, Ayaz, Abbas)
| | - Cheryl A Abbas
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA (Drs Sarwat, Paluy, Boatwright, Browne, Ayaz, Abbas)
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine and the Texas Heart Institute, Houston, TX, USA (Dr Ballantyne)
| |
Collapse
|
4
|
Volgman AS, Koschinsky ML, Mehta A, Rosenson RS. Genetics and Pathophysiological Mechanisms of Lipoprotein(a)-Associated Cardiovascular Risk. J Am Heart Assoc 2024; 13:e033654. [PMID: 38879448 PMCID: PMC11255763 DOI: 10.1161/jaha.123.033654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Abstract
Elevated lipoprotein(a) is a genetically transmitted codominant trait that is an independent risk driver for cardiovascular disease. Lipoprotein(a) concentration is heavily influenced by genetic factors, including LPA kringle IV-2 domain size, single-nucleotide polymorphisms, and interleukin-1 genotypes. Apolipoprotein(a) is encoded by the LPA gene and contains 10 subtypes with a variable number of copies of kringle -2, resulting in >40 different apolipoprotein(a) isoform sizes. Genetic loci beyond LPA, such as APOE and APOH, have been shown to impact lipoprotein(a) levels. Lipoprotein(a) concentrations are generally 5% to 10% higher in women than men, and there is up to a 3-fold difference in median lipoprotein(a) concentrations between racial and ethnic populations. Nongenetic factors, including menopause, diet, and renal function, may also impact lipoprotein(a) concentration. Lipoprotein(a) levels are also influenced by inflammation since the LPA promoter contains an interleukin-6 response element; interleukin-6 released during the inflammatory response results in transient increases in plasma lipoprotein(a) levels. Screening can identify elevated lipoprotein(a) levels and facilitate intensive risk factor management. Several investigational, RNA-targeted agents have shown promising lipoprotein(a)-lowering effects in clinical studies, and large-scale lipoprotein(a) testing will be fundamental to identifying eligible patients should these agents become available. Lipoprotein(a) testing requires routine, nonfasting blood draws, making it convenient for patients. Herein, we discuss the genetic determinants of lipoprotein(a) levels, explore the pathophysiological mechanisms underlying the association between lipoprotein(a) and cardiovascular disease, and provide practical guidance for lipoprotein(a) testing.
Collapse
Affiliation(s)
| | - Marlys L. Koschinsky
- Robarts Research Institute, Schulich School of Medicine and DentistryWestern UniversityLondonONCanada
| | | | - Robert S. Rosenson
- Metabolism and Lipids Program, Mount Sinai Fuster Heart HospitalIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| |
Collapse
|
5
|
Tsegaselassie W, Jian Y, Berhanu GG, Tianyuan L, April M, Tali E, Fasil TA, Timothy TA, Jordana C, Marguerite IR, Robert SM, Michael VW, Kristine Y, Myriam F, Donald LJM, Mario S, Daichi S, Yuichiro Y, Paul M, Adam B. Associations of cardiometabolic polygenic risk scores with cardiovascular disease in African Americans. RESEARCH SQUARE 2023:rs.3.rs-3228815. [PMID: 37693576 PMCID: PMC10491340 DOI: 10.21203/rs.3.rs-3228815/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Background Cardiovascular disease (CVD) is a complex disease, and genetic factors contribute individually or cumulatively to CVD risk. While African American women and men are disproportionately affected by CVD, their lack of representation in genomic investigations may widen disparities in health. We investigated the associations of cardiometabolic polygenic risk scores (PRSs) with CVD risk in African Americans. Methods We used the Jackson Heart Study, a prospective cohort study of CVD in African American adults and the predicted atherosclerotic cardiovascular disease (ASCVD) 10-year risk. We included 40-79 years old adults without a history of coronary heart disease (CHD) or stroke at baseline. We derived genome-wide PRSs for systolic blood pressure (SBP), diastolic blood pressure (DBP), total cholesterol, LDL cholesterol, hemoglobin A1c (HbA1c), triglycerides, and C-reactive protein (CRP) separately for each of the participants, using African-origin UK Biobank participants' genome-wide association summary statistics. We estimated the associations between PRSs and 10-year predicted ASCVD risk adjusting for age, sex, study visit date, and genetic ancestry using linear and logistic regression models. Results Participants (n=2,077) were 63% female and 66% never-smokers. They had mean (SD) 56 (10) years of age, 127.8 (16.3) mmHg SBP, 76.3 (8.7) mmHg DBP, 200.4 (40.2) mg/dL total cholesterol, 51.7 (14.7) mg/dL HDL cholesterol, 127.2 (36.7) mg/dL LDL cholesterol, 6.0 (1.3) mmol/mol HbA1c, 108.9 (81.7) mg/dL triglycerides and 0.53 (1.1) CRP. Their median (interquartile range) predicted 10-year predicted ASCVD risk was 8.0 (4.0-15.0). Participants in the >75th percentile for HbA1c PRS had 1.42 percentage-point greater predicted 10-year ASCVD risk (1.42 [95% CI: 0.58-2.26]) and higher odds of ≥10% predicted 10-year ASCVD risk (OR: 1.46 [95% CI: 1.03-2.07]) compared with those in the <25th percentile for HbA1c PRS. Participants in the >75th percentile for SBP PRS had higher odds of ≥10% predicted 10-year ASCVD risk (OR: 1.52 [95% CI: 1.07-2.15]) compared with those in the <25th percentile for SBP PRS. Conclusion Among 40-79 years old African Americans without CHD and stroke, higher PRSs for HbA1c and SBP were associated with CVD risk. PRSs may help stratify individuals based on their clinical risk factors for CVD early prevention and clinical management.
Collapse
Affiliation(s)
| | | | | | - Lu Tianyuan
- Lady Davis Institute for Medical Research, Jewish General Hospital
| | | | | | | | | | | | | | | | | | | | | | | | - Sims Mario
- University of Mississippi Medical Center
| | | | | | | | | |
Collapse
|
6
|
Maloberti A, Fabbri S, Colombo V, Gualini E, Monticelli M, Daus F, Busti A, Galasso M, De Censi L, Algeri M, Merlini PA, Giannattasio C. Lipoprotein(a): Cardiovascular Disease, Aortic Stenosis and New Therapeutic Option. Int J Mol Sci 2022; 24:ijms24010170. [PMID: 36613613 PMCID: PMC9820656 DOI: 10.3390/ijms24010170] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/23/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a chronic and progressive inflammatory process beginning early in life with late clinical manifestation. This slow pathological trend underlines the importance to early identify high-risk patients and to treat intensively risk factors to prevent the onset and/or the progression of atherosclerotic lesions. In addition to the common Cardiovascular (CV) risk factors, new markers able to increase the risk of CV disease have been identified. Among them, high levels of Lipoprotein(a)-Lp(a)-lead to very high risk of future CV diseases; this relationship has been well demonstrated in epidemiological, mendelian randomization and genome-wide association studies as well as in meta-analyses. Recently, new aspects have been identified, such as its association with aortic stenosis. Although till recent years it has been considered an unmodifiable risk factor, specific drugs have been developed with a strong efficacy in reducing the circulating levels of Lp(a) and their capacity to reduce subsequent CV events is under testing in ongoing trials. In this paper we will review all these aspects: from the synthesis, clearance and measurement of Lp(a), through the findings that examine its association with CV diseases and aortic stenosis to the new therapeutic options that will be available in the next years.
Collapse
Affiliation(s)
- Alessandro Maloberti
- Cardiology 4, Cardio Center A. De Gasperis, ASST GOM Niguarda, 20162 Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
- Correspondence: ; Tel.: +39-02-644-478-55; Fax: +39-02-644-425-66
| | - Saverio Fabbri
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Valentina Colombo
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Elena Gualini
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | | | - Francesca Daus
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Andrea Busti
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Michele Galasso
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Lorenzo De Censi
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| | - Michela Algeri
- Cardiology 4, Cardio Center A. De Gasperis, ASST GOM Niguarda, 20162 Milan, Italy
| | | | - Cristina Giannattasio
- Cardiology 4, Cardio Center A. De Gasperis, ASST GOM Niguarda, 20162 Milan, Italy
- School of Medicine and Surgery, Milano-Bicocca University, 20126 Milan, Italy
| |
Collapse
|
7
|
Fedorova L, Khrunin A, Khvorykh G, Lim J, Thornton N, Mulyar OA, Limborska S, Fedorov A. Analysis of Common SNPs across Continents Reveals Major Genomic Differences between Human Populations. Genes (Basel) 2022; 13:genes13081472. [PMID: 36011383 PMCID: PMC9408407 DOI: 10.3390/genes13081472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/12/2022] [Accepted: 08/17/2022] [Indexed: 12/03/2022] Open
Abstract
Common alleles tend to be more ancient than rare alleles. These common SNPs appeared thousands of years ago and reflect intricate human evolution including various adaptations, admixtures, and migration events. Eighty-four thousand abundant region-specific alleles (ARSAs) that are common in one continent but absent in the rest of the world have been characterized by processing 3100 genomes from 230 populations. Also computed were 17,446 polymorphic sites with regional absence of common alleles (RACAs), which are widespread globally but absent in one region. A majority of these region-specific SNPs were found in Africa. America has the second greatest number of ARSAs (3348) and is even ahead of Europe (1911). Surprisingly, East Asia has the highest number of RACAs (10,524) and the lowest number of ARSAs (362). ARSAs and RACAs have distinct compositions of ancestral versus derived alleles in different geographical regions, reflecting their unique evolution. Genes associated with ARSA and RACA SNPs were identified and their functions were analyzed. The core 100 genes shared by multiple populations and associated with region-specific natural selection were examined. The largest part of them (42%) are related to the nervous system. ARSA and RACA SNPs are important for both association and human evolution studies.
Collapse
Affiliation(s)
| | - Andrey Khrunin
- Institute of Molecular Genetics of National Research Centre, “Kurchatov Institute”, 123182 Moscow, Russia
| | - Gennady Khvorykh
- Institute of Molecular Genetics of National Research Centre, “Kurchatov Institute”, 123182 Moscow, Russia
| | - Jan Lim
- CRI Genetics LLC, Santa Monica, CA 90404, USA
| | | | | | - Svetlana Limborska
- Institute of Molecular Genetics of National Research Centre, “Kurchatov Institute”, 123182 Moscow, Russia
| | - Alexei Fedorov
- CRI Genetics LLC, Santa Monica, CA 90404, USA
- Department of Medicine, University of Toledo, Toledo, OH 43606, USA
- Correspondence: ; Tel.: +1-419-383-5270
| |
Collapse
|
8
|
Lipoprotein(a) beyond the kringle IV repeat polymorphism: The complexity of genetic variation in the LPA gene. Atherosclerosis 2022; 349:17-35. [PMID: 35606073 PMCID: PMC7613587 DOI: 10.1016/j.atherosclerosis.2022.04.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 02/23/2022] [Accepted: 04/01/2022] [Indexed: 12/24/2022]
Abstract
High lipoprotein(a) [Lp(a)] concentrations are one of the most important genetically determined risk factors for cardiovascular disease. Lp(a) concentrations are an enigmatic trait largely controlled by one single gene (LPA) that contains a complex interplay of several genetic elements with many surprising effects discussed in this review. A hypervariable coding copy number variation (the kringle IV type-2 repeat, KIV-2) generates >40 apolipoprotein(a) protein isoforms and determines the median Lp(a) concentrations. Carriers of small isoforms with up to 22 kringle IV domains have median Lp(a) concentrations up to 5 times higher than those with large isoforms (>22 kringle IV domains). The effect of the apo(a) isoforms are, however, modified by many functional single nucleotide polymorphisms (SNPs) distributed over the complete range of allele frequencies (<0.1% to >20%) with very pronounced effects on Lp(a) concentrations. A complex interaction is present between the apo (a) isoforms and LPA SNPs, with isoforms partially masking the effect of functional SNPs and, vice versa, SNPs lowering the Lp(a) concentrations of affected isoforms. This picture is further complicated by SNP-SNP interactions, a poorly understood role of other polymorphisms such as short tandem repeats and linkage structures that are poorly captured by common R2 values. A further layer of complexity derives from recent findings that several functional SNPs are located in the KIV-2 repeat and are thus not accessible to conventional sequencing and genotyping technologies. A critical impact of the ancestry on correlation structures and baseline Lp(a) values becomes increasingly evident. This review provides a comprehensive overview on the complex genetic architecture of the Lp(a) concentrations in plasma, a field that has made tremendous progress with the introduction of new technologies. Understanding the genetics of Lp(a) might be a key to many mysteries of Lp(a) and booster new ideas on the metabolism of Lp(a) and possible interventional targets.
Collapse
|
9
|
Chemello K, Blom DJ, Marais AD, Lambert G, Blanchard V. Genetic and Mechanistic Insights into the Modulation of Circulating Lipoprotein (a) Concentration by Apolipoprotein E Isoforms. Curr Atheroscler Rep 2022; 24:399-405. [PMID: 35355214 DOI: 10.1007/s11883-022-01016-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2022] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW Lipoprotein (a) [Lp(a)] is a highly atherogenic lipoprotein species. A unique feature of Lp(a) is the strong genetic determination of its concentration. The LPA gene is responsible for up to 90% of the variance in Lp(a), but other genes also have an impact. RECENT FINDINGS Genome-wide associations studies indicate that the APOE gene, encoding apolipoprotein E (apoE), is the second most important locus modulating Lp(a) concentrations. Population studies clearly show that carriers of the apoE2 variant (ε2) display reduced Lp(a) levels, the lowest concentrations being observed in ε2/ε2 homozygotes. This genotype can lead predisposed adults to develop dysbetalipoproteinemia, a lipid disorder characterized by sharp elevations in cholesterol and triglycerides. However, dysbetalipoproteinemia does not significantly modulate circulating Lp(a). Mechanistically, apoE appears to impair the production but not the catabolism of Lp(a). These observations underline the complexity of Lp(a) metabolism and provide key insights into the pathways governing Lp(a) synthesis and secretion.
Collapse
Affiliation(s)
- Kévin Chemello
- Laboratoire Inserm, UMR 1188 DéTROI, Université de La Réunion, 2 Rue Maxime Rivière, 97490, Sainte Clotilde, France
| | - Dirk J Blom
- Division of Lipidology and Cape Heart Institute, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - A David Marais
- Division of Chemical Pathology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Gilles Lambert
- Laboratoire Inserm, UMR 1188 DéTROI, Université de La Réunion, 2 Rue Maxime Rivière, 97490, Sainte Clotilde, France.
| | - Valentin Blanchard
- Laboratoire Inserm, UMR 1188 DéTROI, Université de La Réunion, 2 Rue Maxime Rivière, 97490, Sainte Clotilde, France.,Departments of Medicine, Centre for Heart Lung Innovation, Providence Healthcare Research Institute, St. Paul's Hospital, University of British Columbia, Vancouver, Canada
| |
Collapse
|
10
|
Sun Q, Graff M, Rowland B, Wen J, Huang L, Miller-Fleming TW, Haessler J, Preuss MH, Chai JF, Lee MP, Avery CL, Cheng CY, Franceschini N, Sim X, Cox NJ, Kooperberg C, North KE, Li Y, Raffield LM. Analyses of biomarker traits in diverse UK biobank participants identify associations missed by European-centric analysis strategies. J Hum Genet 2022; 67:87-93. [PMID: 34376796 PMCID: PMC8792153 DOI: 10.1038/s10038-021-00968-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/09/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022]
Abstract
Despite the dramatic underrepresentation of non-European populations in human genetics studies, researchers continue to exclude participants of non-European ancestry, as well as variants rare in European populations, even when these data are available. This practice perpetuates existing research disparities and can lead to important and large effect size associations being missed. Here, we conducted genome-wide association studies (GWAS) of 31 serum and urine biomarker quantitative traits in African (n = 9354), East Asian (n = 2559), and South Asian (n = 9823) ancestry UK Biobank (UKBB) participants. We adjusted for all known GWAS catalog variants for each trait, as well as novel signals identified in a recent European ancestry-focused analysis of UKBB participants. We identify 7 novel signals in African ancestry and 2 novel signals in South Asian ancestry participants (p < 1.61E-10). Many of these signals are highly plausible, including a cis pQTL for the gene encoding gamma-glutamyl transferase and PIEZO1 and G6PD variants with impacts on HbA1c through likely erythrocytic mechanisms. This work illustrates the importance of using the genetic data we already have in diverse populations, with novel discoveries possible in even modest sample sizes.
Collapse
Affiliation(s)
- Quan Sun
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Misa Graff
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Bryce Rowland
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Jia Wen
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Le Huang
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Tyne W Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jeffrey Haessler
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jin-Fang Chai
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Moa P Lee
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Christy L Avery
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nora Franceschini
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Nancy J Cox
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Kari E North
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
- Carolina Center of Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Yun Li
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
- Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
11
|
Iranzo-Tatay C, Hervas-Marin D, Rojo-Bofill LM, Garcia D, Vaz-Leal FJ, Calabria I, Beato-Fernandez L, Oltra S, Sandoval J, Rojo-Moreno L. Genome-wide DNA methylation profiling in anorexia nervosa discordant identical twins. Transl Psychiatry 2022; 12:15. [PMID: 35013117 PMCID: PMC8748827 DOI: 10.1038/s41398-021-01776-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 11/18/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022] Open
Abstract
Up until now, no study has looked specifically at epigenomic landscapes throughout twin samples, discordant for Anorexia nervosa (AN). Our goal was to find evidence to confirm the hypothesis that epigenetic variations play a key role in the aetiology of AN. In this study, we quantified genome-wide patterns of DNA methylation using the Infinium Human DNA Methylation EPIC BeadChip array ("850 K") in DNA samples isolated from whole blood collected from a group of 7 monozygotic twin pairs discordant for AN. Results were then validated performing a genome-wide DNA methylation profiling using DNA extracted from whole blood of a group of non-family-related AN patients and a group of healthy controls. Our first analysis using the twin sample revealed 9 CpGs associated to a gene. The validation analysis showed two statistically significant CpGs with the rank regression method related to two genes associated to metabolic traits, PPP2R2C and CHST1. When doing beta regression, 6 of them showed statistically significant differences, including 3 CpGs associated to genes JAM3, UBAP2L and SYNJ2. Finally, the overall pattern of results shows genetic links to phenotypes which the literature has constantly related to AN, including metabolic and psychological traits. The genes PPP2R2C and CHST1 have both been linked to the metabolic traits type 2 diabetes through GWAS studies. The genes UBAP2L and SYNJ2 have been related to other psychiatric comorbidity.
Collapse
Affiliation(s)
- C Iranzo-Tatay
- Psychiatry Service, Hospital la Fe, Valencia, Spain
- Department of Psychiatry, Medicine School, University of Valencia, Valencia, Spain
| | - D Hervas-Marin
- Department of Applied Statistics and Operational Research and Quality, Universitat Politècnica de València, Valencia, Spain
| | | | - D Garcia
- Epigenomics Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - F J Vaz-Leal
- Department of Psychiatry, Medicine School, University of Extremadura, Badajoz, Spain
| | - I Calabria
- Epigenomics Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - L Beato-Fernandez
- Eating Disorders and Children's Psychiatry Department, Hospital General, Ciudad Real, Spain
| | - S Oltra
- Genetics and Prenatal Diagnosis Unit, Hospital La fe, Valencia, Spain
| | - J Sandoval
- Epigenomics Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.
- Biomarkers and Precision Medicine Unit (UByMP), Instituto de Investigación Sanitaria La Fe, Valencia, Spain.
| | - L Rojo-Moreno
- Psychiatry Service, Hospital la Fe, Valencia, Spain
- Department of Psychiatry, Medicine School, University of Valencia, Valencia, Spain
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| |
Collapse
|
12
|
Bensenor I, Padilha K, Lima IR, Santos RD, Lambert G, Ramin-Mangata S, Bittencourt MS, Goulart AC, Santos IS, Mill JG, Krieger JE, Lotufo PA, Pereira AC. Genome-Wide Association of Proprotein Convertase Subtilisin/Kexin Type 9 Plasma Levels in the ELSA-Brasil Study. Front Genet 2021; 12:728526. [PMID: 34659352 PMCID: PMC8514075 DOI: 10.3389/fgene.2021.728526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/16/2021] [Indexed: 11/15/2022] Open
Abstract
Pharmacological inhibition of PCSK9 (proprotein convertase subtilisin/kexin type 9) is an established therapeutic option to treat hypercholesterolemia, and plasma PCSK9 levels have been implicated in cardiovascular disease incidence. A number of genetic variants within the PCSK9 gene locus have been shown to modulate PCSK9 levels, but these only explain a very small percentage of the overall PCSK9 interindividual variation. Here we present data on the genetic association structure between PCSK9 levels and genom-wide genetic variation in a healthy sample from the general population. We performed a genome-wide association study of plasma PCSK9 levels in a sample of Brazilian individuals enrolled in the Estudo Longitudinal de Saude do Adulto cohort (n=810). Enrolled individuals were free from cardiovascular disease, diabetes and were not under lipid-lowering medication. Genome-wide genotyping was conducted using the Axiom_PMRA.r3 array, and imputation was performed using the TOPMED multi-ancestry sample panel as reference. Total PCSK9 plasma concentrations were determined using the Quantikine SPC900 ELISA kit. We observed two genome-wide significant loci and seven loci that reached the pre-defined value of p threshold of 1×10−6. Significant variants were near KCNA5 and KCNA1, and LINC00353. Genetic variation at the PCSK9 locus was able to explain approximately 4% of the overall interindividual variations in PCSK9 levels. Colocalization analysis using eQTL data suggested RWDD3, ATXN7L1, KCNA1, and FAM177A1 to be potential mediators of some of the observed associations. Our results suggest that PCSK9 levels may be modulated by trans genetic variation outside of the PCSK9 gene and this may have clinical implications. Understanding both environmental and genetic predictors of PCSK9 levels may help identify new targets for cardiovascular disease treatment and contribute to a better assessment of the benefits of long-term PCSK9 inhibition.
Collapse
Affiliation(s)
- Isabela Bensenor
- Center for Clinical and Epidemiologic Research, University of São Paulo, São Paulo, Brazil
| | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Isabella Ramos Lima
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Raul Dias Santos
- Lipid Clinic, Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Gilles Lambert
- Inserm UMR 1188 DéTROI, Université La Réunion, Sainte Clotilde, France
| | | | - Marcio S Bittencourt
- Lipid Clinic, Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Alessandra C Goulart
- Center for Clinical and Epidemiologic Research, University of São Paulo, São Paulo, Brazil
| | - Itamar S Santos
- Center for Clinical and Epidemiologic Research, University of São Paulo, São Paulo, Brazil
| | - Jose G Mill
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Brazil
| | - Jose E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Paulo A Lotufo
- Center for Clinical and Epidemiologic Research, University of São Paulo, São Paulo, Brazil
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| |
Collapse
|
13
|
Prokopenko D, Morgan SL, Mullin K, Hofmann O, Chapman B, Kirchner R, Amberkar S, Wohlers I, Lange C, Hide W, Bertram L, Tanzi RE. Whole-genome sequencing reveals new Alzheimer's disease-associated rare variants in loci related to synaptic function and neuronal development. Alzheimers Dement 2021; 17:1509-1527. [PMID: 33797837 PMCID: PMC8519060 DOI: 10.1002/alz.12319] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Genome-wide association studies have led to numerous genetic loci associated with Alzheimer's disease (AD). Whole-genome sequencing (WGS) now permits genome-wide analyses to identify rare variants contributing to AD risk. METHODS We performed single-variant and spatial clustering-based testing on rare variants (minor allele frequency [MAF] ≤1%) in a family-based WGS-based association study of 2247 subjects from 605 multiplex AD families, followed by replication in 1669 unrelated individuals. RESULTS We identified 13 new AD candidate loci that yielded consistent rare-variant signals in discovery and replication cohorts (4 from single-variant, 9 from spatial-clustering), implicating these genes: FNBP1L, SEL1L, LINC00298, PRKCH, C15ORF41, C2CD3, KIF2A, APC, LHX9, NALCN, CTNNA2, SYTL3, and CLSTN2. DISCUSSION Downstream analyses of these novel loci highlight synaptic function, in contrast to common AD-associated variants, which implicate innate immunity and amyloid processing. These loci have not been associated previously with AD, emphasizing the ability of WGS to identify AD-associated rare variants, particularly outside of the exome.
Collapse
Affiliation(s)
- Dmitry Prokopenko
- Genetics and Aging Research Unit and The Henry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| | - Sarah L. Morgan
- Department of NeuroscienceSheffield Institute for Translational NeurosciencesUniversity of SheffieldSheffieldUK
- Department of PathologyBeth Israel Deaconess Medical Center330 Brookline AvenueBostonMassachusettsUSA
| | - Kristina Mullin
- Genetics and Aging Research Unit and The Henry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Oliver Hofmann
- Department of Clinical PathologyUniversity of MelbourneMelbourneVICAustralia
| | - Brad Chapman
- Bioinformatics Core, Harvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Rory Kirchner
- Bioinformatics Core, Harvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | | | - Sandeep Amberkar
- Department of NeuroscienceSheffield Institute for Translational NeurosciencesUniversity of SheffieldSheffieldUK
| | - Inken Wohlers
- Lübeck Interdisciplinary Platform for Genome AnalyticsInstitutes of Neurogenetics and CardiogeneticsUniversity of LübeckLübeckGermany
| | - Christoph Lange
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Winston Hide
- Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeuroscienceSheffield Institute for Translational NeurosciencesUniversity of SheffieldSheffieldUK
- Department of PathologyBeth Israel Deaconess Medical Center330 Brookline AvenueBostonMassachusettsUSA
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome AnalyticsInstitutes of Neurogenetics and CardiogeneticsUniversity of LübeckLübeckGermany
- Department of PsychologyUniversity of OsloOsloNorway
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit and The Henry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General HospitalBostonMassachusettsUSA
- Harvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
14
|
Abstract
Lipoprotein(a) [Lp(a)] is an atherogenic lipoprotein with a strong genetic regulation. Up to 90% of the concentrations are explained by a single gene, the LPA gene. The concentrations show a several-hundred-fold interindividual variability ranging from less than 0.1 mg/dL to more than 300 mg/dL. Lp(a) plasma concentrations above 30 mg/dL and even more above 50 mg/dL are associated with an increased risk for cardiovascular disease including myocardial infarction, stroke, aortic valve stenosis, heart failure, peripheral arterial disease, and all-cause mortality. Since concentrations above 50 mg/dL are observed in roughly 20% of the Caucasian population and in an even higher frequency in African-American and Asian-Indian ethnicities, it can be assumed that Lp(a) is one of the most important genetically determined risk factors for cardiovascular disease.Carriers of genetic variants that are associated with high Lp(a) concentrations have a markedly increased risk for cardiovascular events. Studies that used these genetic variants as a genetic instrument to support a causal role for Lp(a) as a cardiovascular risk factor are called Mendelian randomization studies. The principle of this type of studies has been introduced and tested for the first time ever with Lp(a) and its genetic determinants.There are currently no approved pharmacologic therapies that specifically target Lp(a) concentrations. However, some therapies that target primarily LDL cholesterol have also an influence on Lp(a) concentrations. These are mainly PCSK9 inhibitors that lower LDL cholesterol by 60% and Lp(a) by 25-30%. Furthermore, lipoprotein apheresis lowers both, Lp(a) and LDL cholesterol, by about 60-70%. Some sophisticated study designs and statistical analyses provided support that lowering Lp(a) by these therapies also lowers cardiovascular events on top of the effect caused by lowering LDL cholesterol, although this was not the main target of the therapy. Currently, new therapies targeting RNA such as antisense oligonucleotides (ASO) or small interfering RNA (siRNA) against apolipoprotein(a), the main protein of the Lp(a) particle, are under examination and lower Lp(a) concentrations up to 90%. Since these therapies specifically lower Lp(a) concentrations without influencing other lipoproteins, they will serve the last piece of the puzzle whether a decrease of Lp(a) results also in a decrease of cardiovascular events.
Collapse
|
15
|
Said MA, Yeung MW, van de Vegte YJ, Benjamins JW, Dullaart RPF, Ruotsalainen S, Ripatti S, Natarajan P, Juarez-Orozco LE, Verweij N, van der Harst P. Genome-Wide Association Study and Identification of a Protective Missense Variant on Lipoprotein(a) Concentration: Protective Missense Variant on Lipoprotein(a) Concentration-Brief Report. Arterioscler Thromb Vasc Biol 2021; 41:1792-1800. [PMID: 33730874 DOI: 10.1161/atvbaha.120.315300] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [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)
- M Abdullah Said
- Department of Cardiology (M.A.S., M.W.Y., Y.J.v.d.V., J.W.B., L.E.J.-O., N.V., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Ming Wai Yeung
- Department of Cardiology (M.A.S., M.W.Y., Y.J.v.d.V., J.W.B., L.E.J.-O., N.V., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Yordi J van de Vegte
- Department of Cardiology (M.A.S., M.W.Y., Y.J.v.d.V., J.W.B., L.E.J.-O., N.V., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Jan Walter Benjamins
- Department of Cardiology (M.A.S., M.W.Y., Y.J.v.d.V., J.W.B., L.E.J.-O., N.V., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology (R.P.F.D.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Sanni Ruotsalainen
- Institute for Molecular Medicine Finland HiLIFE (S. Ruotsalainen, S. Ripatti), University of Helsinki, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland HiLIFE (S. Ruotsalainen, S. Ripatti), University of Helsinki, Finland.,Department of Public Health (S. Ripatti), University of Helsinki, Finland.,Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA (S. Ripatti, P.N.)
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA (S. Ripatti, P.N.).,Department of Medicine, Harvard Medical School, Boston, MA (P.N.).,Cardiovascular Research Center, Massachusetts General Hospital, Boston (P.N.)
| | - Luis Eduardo Juarez-Orozco
- Department of Cardiology (M.A.S., M.W.Y., Y.J.v.d.V., J.W.B., L.E.J.-O., N.V., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands.,Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, the Netherlands (L.E.J.-O., P.v.d.H.)
| | - Niek Verweij
- Department of Cardiology (M.A.S., M.W.Y., Y.J.v.d.V., J.W.B., L.E.J.-O., N.V., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - P van der Harst
- Department of Cardiology (M.A.S., M.W.Y., Y.J.v.d.V., J.W.B., L.E.J.-O., N.V., P.v.d.H.), University Medical Center Groningen, University of Groningen, the Netherlands.,Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, University of Utrecht, the Netherlands (L.E.J.-O., P.v.d.H.)
| |
Collapse
|
16
|
Page MM, Watts GF. Contemporary perspectives on the genetics and clinical use of lipoprotein(a) in preventive cardiology. Curr Opin Cardiol 2021; 36:272-280. [PMID: 33741767 DOI: 10.1097/hco.0000000000000842] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW The pathogenicity of lipoprotein(a) [Lp(a)] as a risk factor for atherosclerotic cardiovascular disease (ASCVD) is well evidenced and recognized by international consensus-based guidelines. However, the measurement of Lp(a) is not routine clinical practice. Therapeutic agents targeting Lp(a) are now progressing through randomised clinical trials, and it is timely for clinicians to familiarize themselves with this complex and enigmatic lipoprotein particle. RECENT FINDINGS Recent developments in the understanding of genetic influences on the structure, plasma concentration and atherogenicity of Lp(a) have contextualized its clinical relevance. Mendelian randomization studies have enabled estimation of the contribution of Lp(a) to ASCVD risk. Genotyping individual patients with respect to Lp(a)-raising single nucleotide polymorphisms predicts ASCVD, but has not yet been shown to add value beyond the measurement of Lp(a) plasma concentrations, which should be done by Lp(a) isoform-independent assays capable of reporting in molar concentrations. Contemporary gene-silencing technology underpins small interfering RNA and antisense oligonucleotides, which are emerging as the leading Lp(a)-lowering therapeutic agents. The degree of Lp(a)-lowering required to achieve meaningful reductions in ASCVD risk has been estimated by Mendelian randomization, providing conceptual support. SUMMARY Measurement of Lp(a) in the clinical setting contributes to the assessment of ASCVD risk, and will become more important with the advent of specific Lp(a)-lowering therapies. Knowledge of an individual patient's genetic predisposition to increased Lp(a) appears to impart little or not additional clinical value beyond Lp(a) particle concentration.
Collapse
Affiliation(s)
- Michael M Page
- School of Medicine, University of Western Australia, Crawley
- Western Diagnostic Pathology
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Crawley
- Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
17
|
Hoekstra M, Chen HY, Rong J, Dufresne L, Yao J, Guo X, Tsai MY, Tsimikas S, Post WS, Vasan RS, Rotter JI, Larson MG, Thanassoulis G, Engert JC. Genome-Wide Association Study Highlights APOH as a Novel Locus for Lipoprotein(a) Levels-Brief Report. Arterioscler Thromb Vasc Biol 2021; 41:458-464. [PMID: 33115273 PMCID: PMC7769958 DOI: 10.1161/atvbaha.120.314965] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Lp(a) (lipoprotein[a]) is an independent risk factor for cardiovascular diseases and plasma levels are primarily determined by variation at the LPA locus. We performed a genome-wide association study in the UK Biobank to determine whether additional loci influence Lp(a) levels. Approach and Results: We included 293 274 White British individuals in the discovery analysis. Approximately 93 095 623 variants were tested for association with natural log-transformed Lp(a) levels using linear regression models adjusted for age, sex, genotype batch, and 20 principal components of genetic ancestry. After quality control, 131 independent variants were associated at genome-wide significance (P≤5×10-8). In addition to validating previous associations at LPA, APOE, and CETP, we identified a novel variant at the APOH locus, encoding β2GPI (beta2-glycoprotein I). The APOH variant rs8178824 was associated with increased Lp(a) levels (β [95% CI] [ln nmol/L], 0.064 [0.047-0.081]; P=2.8×10-13) and demonstrated a stronger effect after adjustment for variation at the LPA locus (β [95% CI] [ln nmol/L], 0.089 [0.076-0.10]; P=3.8×10-42). This association was replicated in a meta-analysis of 5465 European-ancestry individuals from the Framingham Offspring Study and Multi-Ethnic Study of Atherosclerosis (β [95% CI] [ln mg/dL], 0.16 [0.044-0.28]; P=0.0071). CONCLUSIONS In a large-scale genome-wide association study of Lp(a) levels, we identified APOH as a novel locus for Lp(a) in individuals of European ancestry. Additional studies are needed to determine the precise role of β2GPI in influencing Lp(a) levels as well as its potential as a therapeutic target.
Collapse
Affiliation(s)
- Mary Hoekstra
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - Hao Yu Chen
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - Jian Rong
- Boston University’s and NHLBI’s Framingham Heart Study, Boston, Massachusetts
| | - Line Dufresne
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Michael Y. Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota
| | - Sotirios Tsimikas
- Division of Cardiovascular Medicine, Sulpizio Cardiovascular Center, University of California San Diego, La Jolla, California
| | - Wendy S. Post
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, California
| | - Martin G. Larson
- Boston University’s and NHLBI’s Framingham Heart Study, Boston, Massachusetts
| | - George Thanassoulis
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
| | - James C. Engert
- Division of Experimental Medicine, McGill University, Montreal, Quebec
- Preventive and Genomic Cardiology, McGill University Health Centre and Research Institute, Montreal, Quebec
- Department of Human Genetics, McGill University, Montreal, Quebec
| |
Collapse
|
18
|
Prokopenko D, Morgan SL, Mullin K, Hofmann O, Chapman B, Kirchner R, Amberkar S, Wohlers I, Lange C, Hide W, Bertram L, Tanzi RE. Whole-genome sequencing reveals new Alzheimer's disease-associated rare variants in loci related to synaptic function and neuronal development. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.11.03.20225540. [PMID: 33173892 PMCID: PMC7654884 DOI: 10.1101/2020.11.03.20225540] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
INTRODUCTION Genome-wide association studies have led to numerous genetic loci associated with Alzheimer's disease (AD). Whole-genome sequencing (WGS) now permit genome-wide analyses to identify rare variants contributing to AD risk. METHODS We performed single-variant and spatial clustering-based testing on rare variants (minor allele frequency ≤1%) in a family-based WGS-based association study of 2,247 subjects from 605 multiplex AD families, followed by replication in 1,669 unrelated individuals. RESULTS We identified 13 new AD candidate loci that yielded consistent rare-variant signals in discovery and replication cohorts (4 from single-variant, 9 from spatial-clustering), implicating these genes: FNBP1L, SEL1L, LINC00298, PRKCH, C15ORF41, C2CD3, KIF2A, APC, LHX9, NALCN, CTNNA2, SYTL3, CLSTN2. DISCUSSION Downstream analyses of these novel loci highlight synaptic function, in contrast to common AD-associated variants, which implicate innate immunity. These loci have not been previously associated with AD, emphasizing the ability of WGS to identify AD-associated rare variants, particularly outside of coding regions.
Collapse
|
19
|
Han W, Wei Z, Dang R, Guo Y, Zhang H, Geng C, Wang C, Feng Q, Jiang P. Angiotensin-Ⅱ and angiotensin-(1-7) imbalance affects comorbidity of depression and coronary heart disease. Peptides 2020; 131:170353. [PMID: 32599080 DOI: 10.1016/j.peptides.2020.170353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 01/03/2023]
Abstract
A large body of evidence suggests a relationship between depression and coronary heart disease (CHD). Angiotensin-Ⅱ (Ang-Ⅱ) and angiotensin-(1-7) [Ang-(1-7)] are considered to exert biological effects in both conditions. Here, we aimed to determine the role of Ang-Ⅱ and Ang-(1-7) in the occurrence of comorbid depression in patients with CHD. Our study included 214 CHD patients and 100 matched healthy controls. Serum Ang-Ⅱ and Ang-(1-7) levels were assessed by ELISA, and the depression symptoms were evaluated by the nine-item Patient Health Questionnaire (PHQ-9). Linear regression and correlation analyses were used to estimate the associations between PHQ-9 scores and Ang-Ⅱ and Ang-(1-7) serum levels. Six single-nucleotide polymorphisms (SNPs) spanning the angiotensin converting enzyme 2 (ACE2) and MAS1 genes were genotyped. The associations between SNPs and depression risk in CHD patients were examined using logistic regression analysis with adjustment for age and gender. Decreased Ang-(1-7) (P < 0.05) and an elevated Ang-Ⅱ/Ang-(1-7) ratio (P < 0.01) were observed in CHD patients with depression compared to CHD patients without depression. PHQ-9 scores were negatively correlated with Ang-(1-7) level (r=-0.44, P < 0.01) and positively correlated with the Ang-Ⅱ/Ang-(1-7) ratio (r = 0.33, P < 0.05). Furthermore, carriers of risk allele T for CHD with depression had significantly higher PHQ-9 scores (P < 0.05), lower Ang-(1-7) level (P < 0.01), and higher Ang-Ⅱ/Ang-(1-7) ratio (P < 0.05) than those CC carriers. Collectively, our results firstly showed that Ang-(1-7) serum level in CHD patients may protect against comorbid depression. Moreover, the imbalance between Ang-Ⅱ and Ang-(1-7) may contribute to depression in CHD patients.
Collapse
Affiliation(s)
- Wenxiu Han
- Jining First People's Hospital, Jining Medical University, Jining 272000, China
| | - Zhijie Wei
- Jining First People's Hospital, Jining Medical University, Jining 272000, China
| | - Ruili Dang
- Jining First People's Hospital, Jining Medical University, Jining 272000, China
| | - Yujin Guo
- Jining First People's Hospital, Jining Medical University, Jining 272000, China
| | - Hailiang Zhang
- Jining First People's Hospital, Jining Medical University, Jining 272000, China
| | - Chunmei Geng
- Jining First People's Hospital, Jining Medical University, Jining 272000, China
| | - Changshui Wang
- Department of Clinical & Translational Medicine, Jining Life Science Center, Jining 272000, China
| | - Qingyan Feng
- Jining First People's Hospital, Jining Medical University, Jining 272000, China.
| | - Pei Jiang
- Jining First People's Hospital, Jining Medical University, Jining 272000, China.
| |
Collapse
|
20
|
Di Maio S, Grüneis R, Streiter G, Lamina C, Maglione M, Schoenherr S, Öfner D, Thorand B, Peters A, Eckardt KU, Köttgen A, Kronenberg F, Coassin S. Investigation of a nonsense mutation located in the complex KIV-2 copy number variation region of apolipoprotein(a) in 10,910 individuals. Genome Med 2020; 12:74. [PMID: 32825847 PMCID: PMC7442989 DOI: 10.1186/s13073-020-00771-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 08/05/2020] [Indexed: 01/23/2023] Open
Abstract
Background The concentrations of the highly atherogenic lipoprotein(a) [Lp(a)] are mainly genetically determined by the LPA gene locus. However, up to 70% of the coding sequence is located in the complex so-called kringle IV type 2 (KIV-2) copy number variation, a region hardly accessible by common genotyping and sequencing technologies. Despite its size, little is known about genetic variants in this complex region. The R21X variant is a functional variant located in this region, but it has never been analyzed in large cohorts. Methods We typed R21X in 10,910 individuals from three European populations using a newly developed high-throughput allele-specific qPCR assay. R21X allelic location was determined by separating the LPA alleles using pulsed-field gel electrophoresis (PFGE) and typing them separately. Using GWAS data, we identified a proxy SNP located outside of the KIV-2. Linkage disequilibrium was determined both statistically and by long-range haplotyping using PFGE. Worldwide frequencies were determined by reanalyzing the sequencing data of the 1000 Genomes Project with a dedicated pipeline. Results R21X carriers (frequency 0.016–0.021) showed significantly lower mean Lp(a) concentrations (− 11.7 mg/dL [− 15.5; − 7.82], p = 3.39e−32). The variant is located mostly on medium-sized LPA alleles. In the 1000 Genome data, R21X mostly occurs in Europeans and South Asians, is absent in Africans, and shows varying frequencies in South American populations (0 to 0.022). Of note, the best proxy SNP was another LPA null mutation (rs41272114, D′ = 0.958, R2 = 0.281). D′ was very high in all 1000G populations (0.986–0.996), although rs41272114 frequency varies considerably (0–0.182). Co-localization of both null mutations on the same allele was confirmed by PFGE-based long-range haplotyping. Conclusions We performed the largest epidemiological study on an LPA KIV-2 variant so far, showing that it is possible to assess LPA KIV-2 mutations on a large scale. Surprisingly, in all analyzed populations, R21X was located on the same haplotype as the splice mutation rs41272114, creating “double-null” LPA alleles. Despite being a nonsense variant, the R21X status does not provide additional information beyond the rs41272114 genotype. This has important implications for studies using LPA loss-of-function mutations as genetic instruments and emphasizes the complexity of LPA genetics.
Collapse
Affiliation(s)
- Silvia Di Maio
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Schöpfstrasse 41, A-6020, Innsbruck, Austria
| | - Rebecca Grüneis
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Schöpfstrasse 41, A-6020, Innsbruck, Austria
| | - Gertraud Streiter
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Schöpfstrasse 41, A-6020, Innsbruck, Austria
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Schöpfstrasse 41, A-6020, Innsbruck, Austria
| | - Manuel Maglione
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schoenherr
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Schöpfstrasse 41, A-6020, Innsbruck, Austria
| | - Dietmar Öfner
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Thorand
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Neuherberg, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany.,Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Schöpfstrasse 41, A-6020, Innsbruck, Austria
| | - Stefan Coassin
- Institute of Genetic Epidemiology, Department of Genetics and Pharmacology, Medical University of Innsbruck, Schöpfstrasse 41, A-6020, Innsbruck, Austria.
| |
Collapse
|
21
|
Coassin S, Hermann-Kleiter N, Haun M, Wahl S, Wilson R, Paulweber B, Kunze S, Meitinger T, Strauch K, Peters A, Waldenberger M, Kronenberg F, Lamina C. A genome-wide analysis of DNA methylation identifies a novel association signal for Lp(a) concentrations in the LPA promoter. PLoS One 2020; 15:e0232073. [PMID: 32343731 PMCID: PMC7188291 DOI: 10.1371/journal.pone.0232073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/19/2020] [Indexed: 12/24/2022] Open
Abstract
Lipoprotein(a) [Lp(a)] is a major cardiovascular risk factor, which is largely genetically determined by one major gene locus, the LPA gene. Many aspects of the transcriptional regulation of LPA are poorly understood and the role of epigenetics has not been addressed yet. Therefore, we conducted an epigenome-wide analysis of DNA methylation on Lp(a) levels in two population-based studies (total n = 2208). We identified a CpG site in the LPA promoter which was significantly associated with Lp(a) concentrations. Surprisingly, the identified CpG site was found to overlap the SNP rs76735376. We genotyped this SNP de-novo in three studies (total n = 7512). The minor allele of rs76735376 (1.1% minor allele frequency) was associated with increased Lp(a) values (p = 1.01e-59) and explained 3.5% of the variation of Lp(a). Statistical mediation analysis showed that the effect on Lp(a) is rather originating from the base change itself and is not mediated by DNA methylation levels. This finding is supported by eQTL data from 208 liver tissue samples from the GTEx project, which shows a significant association of the rs76735376 minor allele with increased LPA expression. To evaluate, whether the association signal at rs76735376 may actually be derived from a stronger eQTL signal in LD with this SNP, eQTL association results of all correlated SNPs (r2≥0.1) were integrated with genetic association results. This analysis pinpointed to rs10455872 as the potential trigger of the effect of rs76735376. Furthermore, both SNPs coincide with short apo(a) isoforms. Adjusting for both, rs10455872 and the apo(a) isoforms diminished the effect size of rs76735376 to 5.38 mg/dL (p = 0.0463). This indicates that the effect of rs76735376 can be explained by both an independent effect of the SNP and a strong correlation with rs10455872 and apo(a) isoforms.
Collapse
Affiliation(s)
- Stefan Coassin
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Natascha Hermann-Kleiter
- Department of Genetics and Pharmacology, Institute of Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Margot Haun
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Simone Wahl
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Rory Wilson
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Bernhard Paulweber
- First Department of Internal Medicine, Paracelsus Private Medical University, Salzburg, Austria
| | - Sonja Kunze
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Meitinger
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
- Institute of Human Genetics, Technische Universität München, Munich, Germany
- German Research Center for Environmental Health, Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Konstantin Strauch
- German Research Center for Environmental Health, Institute of Genetic Epidemiology, Helmholtz Zentrum München, Neuherberg, Germany
- Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum München–German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Florian Kronenberg
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
22
|
Morgan BM, Brown AN, Deo N, Harrop TWR, Taiaroa G, Mace PD, Wilbanks SM, Merriman TR, Williams MJA, McCormick SPA. Nonsynonymous SNPs in LPA homologous to plasminogen deficiency mutants represent novel null apo(a) alleles. J Lipid Res 2019; 61:432-444. [PMID: 31806727 DOI: 10.1194/jlr.m094540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 11/18/2019] [Indexed: 12/20/2022] Open
Abstract
Plasma lipoprotein (a) [Lp(a)] levels are largely determined by variation in the LPA gene, which codes for apo(a). Genome-wide association studies (GWASs) have identified nonsynonymous variants in LPA that associate with low Lp(a) levels, although their effect on apo(a) function is unknown. We investigated two such variants, R990Q and R1771C, which were present in four null Lp(a) individuals, for structural and functional effects. Sequence alignments showed the R990 and R1771 residues to be highly conserved and homologous to each other and to residues associated with plasminogen deficiency. Structural modeling showed both residues to make several polar contacts with neighboring residues that would be ablated on substitution. Recombinant expression of the WT and R1771C apo(a) in liver and kidney cells showed an abundance of an immature form for both apo(a) proteins. A mature form of apo(a) was only seen with the WT protein. Imaging of the recombinant apo(a) proteins in conjunction with markers of the secretory pathway indicated a poor transit of R1771C into the Golgi. Furthermore, the R1771C mutant displayed a glycosylation pattern consistent with ER, but not Golgi, glycosylation. We conclude that R1771 and the equivalent R990 residue facilitate correct folding of the apo(a) kringle structure and mutations at these positions prevent the proper folding required for full maturation and secretion. To our knowledge, this is the first example of nonsynonymous variants in LPA being causative of a null Lp(a) phenotype.
Collapse
Affiliation(s)
- Benjamin M Morgan
- Department of Biochemistry, School of Biomedical Sciences University of Otago, Dunedin, New Zealand
| | - Aimee N Brown
- Department of Biochemistry, School of Biomedical Sciences University of Otago, Dunedin, New Zealand
| | - Nikita Deo
- Department of Biochemistry, School of Biomedical Sciences University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Tom W R Harrop
- Department of Biochemistry, School of Biomedical Sciences University of Otago, Dunedin, New Zealand
| | - George Taiaroa
- Department of Biochemistry, School of Biomedical Sciences University of Otago, Dunedin, New Zealand
| | - Peter D Mace
- Department of Biochemistry, School of Biomedical Sciences University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Sigurd M Wilbanks
- Department of Biochemistry, School of Biomedical Sciences University of Otago, Dunedin, New Zealand
| | - Tony R Merriman
- Department of Biochemistry, School of Biomedical Sciences University of Otago, Dunedin, New Zealand.,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Michael J A Williams
- Department of Medicine, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | - Sally P A McCormick
- Department of Biochemistry, School of Biomedical Sciences University of Otago, Dunedin, New Zealand .,Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
23
|
Abstract
BACKGROUND Age at menarche and age at natural menopause occur significantly earlier in African American women than in other ethnic groups. African American women also have twice the prevalence of cardiometabolic disorders related to the timing of these reproductive traits. OBJECTIVES The objectives of this integrative review were to (a) summarize the genome-wide association studies of reproductive traits in African American women, (b) identify genes that overlap with reproductive traits and cardiometabolic risk factors in African American women, and (c) propose biological mechanisms explaining the link between reproductive traits and cardiometabolic risk factors. METHODS PubMed was searched for genome-wide association studies of genes associated with reproductive traits in African American women. After extracting and summarizing the primary genes, we examined whether any of the associations with reproductive traits had also been identified with cardiometabolic risk factors in African American women. RESULTS Seven studies met the inclusion criteria. Associations with both reproductive and cardiometabolic traits were reported in or near the following genes: FTO, SEC16B, TMEM18, APOE, PHACTR1, KCNQ1, LDLR, PIK3R1, and RORA. Biological pathways implicated include body weight regulation, vascular homeostasis, and lipid metabolism. DISCUSSION A better understanding of the genetic basis of reproductive traits in African American women may provide insight into the biological mechanisms linking variation in these traits with increased risk for cardiometabolic disorders in this population.
Collapse
|
24
|
Liu Y, Chen S, Li Z, Morrison AC, Boerwinkle E, Lin X. ACAT: A Fast and Powerful p Value Combination Method for Rare-Variant Analysis in Sequencing Studies. Am J Hum Genet 2019; 104:410-421. [PMID: 30849328 PMCID: PMC6407498 DOI: 10.1016/j.ajhg.2019.01.002] [Citation(s) in RCA: 238] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022] Open
Abstract
Set-based analysis that jointly tests the association of variants in a group has emerged as a popular tool for analyzing rare and low-frequency variants in sequencing studies. The existing set-based tests can suffer significant power loss when only a small proportion of variants are causal, and their powers can be sensitive to the number, effect sizes, and effect directions of the causal variants and the choices of weights. Here we propose an aggregated Cauchy association test (ACAT), a general, powerful, and computationally efficient p value combination method for boosting power in sequencing studies. First, by combining variant-level p values, we use ACAT to construct a set-based test (ACAT-V) that is particularly powerful in the presence of only a small number of causal variants in a variant set. Second, by combining different variant-set-level p values, we use ACAT to construct an omnibus test (ACAT-O) that combines the strength of multiple complimentary set-based tests, including the burden test, sequence kernel association test (SKAT), and ACAT-V. Through analysis of extensively simulated data and the whole-genome sequencing data from the Atherosclerosis Risk in Communities (ARIC) study, we demonstrate that ACAT-V complements the SKAT and the burden test, and that ACAT-O has a substantially more robust and higher power than those of the alternative tests.
Collapse
Affiliation(s)
- Yaowu Liu
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Sixing Chen
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Zilin Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Alanna C Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Eric Boerwinkle
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
25
|
Zekavat SM, Ruotsalainen S, Handsaker RE, Alver M, Bloom J, Poterba T, Seed C, Ernst J, Chaffin M, Engreitz J, Peloso GM, Manichaikul A, Yang C, Ryan KA, Fu M, Johnson WC, Tsai M, Budoff M, Vasan RS, Cupples LA, Rotter JI, Rich SS, Post W, Mitchell BD, Correa A, Metspalu A, Wilson JG, Salomaa V, Kellis M, Daly MJ, Neale BM, McCarroll S, Surakka I, Esko T, Ganna A, Ripatti S, Kathiresan S, Natarajan P. Deep coverage whole genome sequences and plasma lipoprotein(a) in individuals of European and African ancestries. Nat Commun 2018; 9:2606. [PMID: 29973585 PMCID: PMC6031652 DOI: 10.1038/s41467-018-04668-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 05/15/2018] [Indexed: 02/06/2023] Open
Abstract
Lipoprotein(a), Lp(a), is a modified low-density lipoprotein particle that contains apolipoprotein(a), encoded by LPA, and is a highly heritable, causal risk factor for cardiovascular diseases that varies in concentrations across ancestries. Here, we use deep-coverage whole genome sequencing in 8392 individuals of European and African ancestry to discover and interpret both single-nucleotide variants and copy number (CN) variation associated with Lp(a). We observe that genetic determinants between Europeans and Africans have several unique determinants. The common variant rs12740374 associated with Lp(a) cholesterol is an eQTL for SORT1 and independent of LDL cholesterol. Observed associations of aggregates of rare non-coding variants are largely explained by LPA structural variation, namely the LPA kringle IV 2 (KIV2)-CN. Finally, we find that LPA risk genotypes confer greater relative risk for incident atherosclerotic cardiovascular diseases compared to directly measured Lp(a), and are significantly associated with measures of subclinical atherosclerosis in African Americans.
Collapse
Affiliation(s)
- Seyedeh M Zekavat
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Yale School of Medicine, New Haven, CT, 06510, USA
- Department of Computational Biology & Bioinformatics, Yale University, New Haven, CT, 06510, USA
| | - Sanni Ruotsalainen
- Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Robert E Handsaker
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Maris Alver
- Department of Biotechnology, Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
- Estonian Genome Center, Tallinn, Estonia
| | - Jonathan Bloom
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Boston, MA, 02142, USA
| | - Timothy Poterba
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Boston, MA, 02142, USA
| | - Cotton Seed
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Boston, MA, 02142, USA
| | - Jason Ernst
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Mark Chaffin
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Jesse Engreitz
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Gina M Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22904, USA
| | - Chaojie Yang
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22904, USA
| | - Kathleen A Ryan
- Program in Personalized and Genomic Medicine, Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Mao Fu
- Program in Personalized and Genomic Medicine, Division of Endocrinology, Diabetes & Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - W Craig Johnson
- Department of Biostatistics, School of Public Health and Community Medicine, University of Washington, Seattle, WA, 98195, USA
| | - Michael Tsai
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Matthew Budoff
- Division of Cardiology, Harbor-UCLA Medical Center, Los Angeles Biomedical Research Institute, Los Angeles, CA, 90509, USA
| | - Ramachandran S Vasan
- NHLBI Framingham Heart Study, Framingham, MA, 20892, USA
- Sections of Preventive medicine and Epidemiology, and cardiovascular medicine, Departments of Medicine and Epidemiology, Boston university Schools of Medicine and Public health, Boston, MA, 02118, USA
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
- NHLBI Framingham Heart Study, Framingham, MA, 20892, USA
| | - Jerome I Rotter
- Departments of Pediatrics and Medicine, The Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, Harbor-UCLA Medical Center, Torrance, CA, 90509, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, 22904, USA
| | - Wendy Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Braxton D Mitchell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | | | - James G Wilson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki, Finland
| | - Manolis Kellis
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, 32 Vassar St, Cambridge, MA, 02139, USA
| | - Mark J Daly
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Boston, MA, 02142, USA
| | - Benjamin M Neale
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Boston, MA, 02142, USA
| | - Steven McCarroll
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Ida Surakka
- Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Tonu Esko
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Estonian Genome Center, Tallinn, Estonia
| | - Andrea Ganna
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Analytic and Translational Genetics Unit, Boston, MA, 02142, USA
| | - Samuli Ripatti
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Institute for Molecular Medicine, University of Helsinki, Helsinki, Finland
- Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sekar Kathiresan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, 02114, USA.
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, 02114, USA.
| |
Collapse
|
26
|
Organic cation transporter 1 (OCT1) modulates multiple cardiometabolic traits through effects on hepatic thiamine content. PLoS Biol 2018; 16:e2002907. [PMID: 29659562 PMCID: PMC5919692 DOI: 10.1371/journal.pbio.2002907] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 04/26/2018] [Accepted: 03/20/2018] [Indexed: 01/07/2023] Open
Abstract
A constellation of metabolic disorders, including obesity, dysregulated lipids, and elevations in blood glucose levels, has been associated with cardiovascular disease and diabetes. Analysis of data from recently published genome-wide association studies (GWAS) demonstrated that reduced-function polymorphisms in the organic cation transporter, OCT1 (SLC22A1), are significantly associated with higher total cholesterol, low-density lipoprotein (LDL) cholesterol, and triglyceride (TG) levels and an increased risk for type 2 diabetes mellitus, yet the mechanism linking OCT1 to these metabolic traits remains puzzling. Here, we show that OCT1, widely characterized as a drug transporter, plays a key role in modulating hepatic glucose and lipid metabolism, potentially by mediating thiamine (vitamin B1) uptake and hence its levels in the liver. Deletion of Oct1 in mice resulted in reduced activity of thiamine-dependent enzymes, including pyruvate dehydrogenase (PDH), which disrupted the hepatic glucose–fatty acid cycle and shifted the source of energy production from glucose to fatty acids, leading to a reduction in glucose utilization, increased gluconeogenesis, and altered lipid metabolism. In turn, these effects resulted in increased total body adiposity and systemic levels of glucose and lipids. Importantly, wild-type mice on thiamine deficient diets (TDs) exhibited impaired glucose metabolism that phenocopied Oct1 deficient mice. Collectively, our study reveals a critical role of hepatic thiamine deficiency through OCT1 deficiency in promoting the metabolic inflexibility that leads to the pathogenesis of cardiometabolic disease. The liver is the major organ for glucose and lipid metabolism; impairment in liver energy metabolism is often found in metabolic disorders. Traditionally, excesses in macronutrients (fat and glucose) are linked to the development of metabolic disorders. Our study provides evidence that imbalances in a micronutrient, vitamin B1 (thiamine), can serve as an etiological cause of lipid and glucose disorders and implicates the organic cation transporter, OCT1, in these disorders. OCT1 is a key determinant of thiamine levels in the liver. In humans, reduced-function polymorphisms of OCT1 significantly associate with high LDL cholesterol levels. Using Oct1 knockout mice, we show that reduced OCT1-mediated thiamine uptake in the liver leads to reduced levels of TPP—the active metabolite of thiamine—and decreased activity of key TPP-dependent enzymes. As a result, a shift from glucose to fatty acid oxidation occurs, leading to imbalances in key metabolic intermediates, alterations in metabolic flux pathways, and disruptions of various metabolic regulatory mechanisms. The extensive characterization of Oct1 knockout mice provides evidence for the molecular mechanisms responsible for various metabolic traits and indicates an important role for imbalances in micronutrients in cardiometabolic disorders.
Collapse
|
27
|
Erhart G, Lamina C, Lehtimäki T, Marques-Vidal P, Kähönen M, Vollenweider P, Raitakari OT, Waeber G, Thorand B, Strauch K, Gieger C, Meitinger T, Peters A, Kronenberg F, Coassin S. Genetic Factors Explain a Major Fraction of the 50% Lower Lipoprotein(a) Concentrations in Finns. Arterioscler Thromb Vasc Biol 2018; 38:1230-1241. [PMID: 29567679 PMCID: PMC5943067 DOI: 10.1161/atvbaha.118.310865] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 02/26/2018] [Indexed: 11/17/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Lp(a) (lipoprotein(a)) concentrations are widely genetically determined by the LPA isoforms and show 5-fold interpopulation differences. Two- to 3-fold differences have been reported even within Europe. Finns represent a distinctive population isolate within Europe and have been repeatedly reported to present lower Lp(a) concentrations than Central Europeans. The significance of this finding was unclear for a long time because of the difficult comparability of Lp(a) assays. Recently, a large standardized study in >50 000 individuals from 7 European populations confirmed this observation but could not provide insights into the causes. Approach and Results— We investigated Lp(a) concentrations, LPA isoforms, and genotypes of established genetic variants affecting Lp(a) concentrations (LPA variants, APOE isoforms, and PCSK9 R46L) in the Finnish YFS (Cardiovascular Risk in Young Finns Study) population (n=2281) and 3 Non-Finnish Central European populations (n=10 003). We observed ≈50% lower Lp(a) concentrations in Finns. The isoform distribution was shifted toward longer isoforms, and the percentage of low-molecular-weight isoform carriers was reduced. Most interestingly, however, Lp(a) was reduced in each single-isoform group. In contrast to the known inverse relationship between LPA isoforms and Lp(a) concentrations, especially very short isoforms presented unexpectedly low Lp(a) concentrations in Finns. The investigated genetic variants, as well as age, sex, and renal function, explained 71.8% of the observed population differences. Conclusions— The population differences in Lp(a) concentrations between Finnish and Central European populations originate not only from a different LPA isoform distribution but suggest the existence of novel functional variation in the small-isoform range.
Collapse
Affiliation(s)
- Gertraud Erhart
- From the Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Austria (G.E., C.L., F.K., S.C.)
| | - Claudia Lamina
- From the Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Austria (G.E., C.L., F.K., S.C.)
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories (T.L.).,Finnish Cardiovascular Research Center (T.L., M.K.)
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Switzerland (P.M.-V., P.V., G.W.)
| | - Mika Kähönen
- Finnish Cardiovascular Research Center (T.L., M.K.).,Department of Clinical Physiology, Tampere University Hospital (M.K.), University of Tampere, Finland
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Switzerland (P.M.-V., P.V., G.W.)
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Finland (O.T.R.).,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Finland (O.T.R.)
| | - Gérard Waeber
- Department of Medicine, Internal Medicine, Lausanne University Hospital, Switzerland (P.M.-V., P.V., G.W.)
| | - Barbara Thorand
- Institute of Epidemiology II (B.T., C.G., A.P.).,German Center for Diabetes Research, Neuherberg, Germany (B.T., A.P.)
| | - Konstantin Strauch
- Institute of Genetic Epidemiology (K.S., C.G.).,Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universität, Munich, Germany (K.S.)
| | - Christian Gieger
- Institute of Epidemiology II (B.T., C.G., A.P.).,Institute of Genetic Epidemiology (K.S., C.G.).,Research Unit of Molecular Epidemiology (C.G.), Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Technische Universität München, Germany (T.M.).,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany (T.M.).,Munich Cluster for Systems Neurology, Germany (T.M.)
| | - Annette Peters
- Institute of Epidemiology II (B.T., C.G., A.P.).,German Center for Diabetes Research, Neuherberg, Germany (B.T., A.P.).,German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance (A.P.)
| | - Florian Kronenberg
- From the Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Austria (G.E., C.L., F.K., S.C.)
| | - Stefan Coassin
- From the Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Austria (G.E., C.L., F.K., S.C.)
| |
Collapse
|
28
|
Page MM, Watts GF. PCSK9 in context: A contemporary review of an important biological target for the prevention and treatment of atherosclerotic cardiovascular disease. Diabetes Obes Metab 2018; 20:270-282. [PMID: 28736830 DOI: 10.1111/dom.13070] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/19/2017] [Accepted: 07/19/2017] [Indexed: 12/16/2022]
Abstract
The identification of the critical role of proprotein convertase subtilisin/kexin type 9 (PCSK9) has rapidly led to the development of PCSK9 inhibition with monoclonal antibodies (mAbs). PCSK9 mAbs are already in limited clinical use and are the subject of major cardiovascular outcomes trials, which, if universally positive, could see much wider clinical application of these agents. Patients with familial hypercholesterolaemia are the most obvious candidates for these drugs, but other patients with elevated cardiovascular risk, statin intolerance or hyperlipoproteinaemia(a) may also benefit. PCSK9 mAbs, administered once or twice monthly, reduce LDL cholesterol levels by 50% to 70%, and appear to be safe and acceptable to patients over at least 2 years of treatment; however, treatment-emergent adverse effects are not always identified in clinical trials, as well-evidenced by statin myopathy. Inclisiran is a promising RNA-based therapy that promotes the degradation of PCSK9 mRNA transcripts and has similar efficacy to mAbs, but with a much longer duration of action. The cost-effectiveness and long-term safety of therapies targeted at inhibiting PCSK9 remain to be demonstrated if they are to be used widely in coronary prevention.
Collapse
Affiliation(s)
- Michael M Page
- Department of Clinical Biochemistry, PathWest Laboratory Medicine, Fiona Stanley Hospital, Perth, Western Australia
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia
| | - Gerald F Watts
- Lipid Disorders Clinic, Department of Cardiology, Royal Perth Hospital, Perth, Western Australia
- School of Medicine, Faculty of Health and Medical Sciences, University of Western Australia, Perth, Western Australia
| |
Collapse
|
29
|
Ellis KL, Boffa MB, Sahebkar A, Koschinsky ML, Watts GF. The renaissance of lipoprotein(a): Brave new world for preventive cardiology? Prog Lipid Res 2017; 68:57-82. [DOI: 10.1016/j.plipres.2017.09.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/01/2017] [Accepted: 09/05/2017] [Indexed: 12/24/2022]
|
30
|
Kritharides L, Nordestgaard BG, Tybjærg-Hansen A, Kamstrup PR, Afzal S. Effect of APOE ε Genotype on Lipoprotein(a) and the Associated Risk of Myocardial Infarction and Aortic Valve Stenosis. J Clin Endocrinol Metab 2017. [PMID: 28651346 DOI: 10.1210/jc.2017-01049] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
CONTEXT APOEε2/3/4 genotypes affect plasma lipoprotein(a); however, the effects of APOE genotypes on the prediction of myocardial infarction and aortic valve stenosis by lipoprotein(a) are unknown. OBJECTIVE We tested the hypothesis that APOEε2/3/4 genotype affects plasma lipoprotein(a), the contribution of plasma apoE levels to this association as well as the associated risk of myocardial infarction and aortic valve stenosis. DESIGN AND OUTCOME MEASURES In 46,615 individuals from the general population, we examined plasma lipoprotein(a), APOE ε2/3/4, and incidence of myocardial infarction (n = 1807) and aortic valve stenosis (n = 345) over 37 years of follow-up (range: 0.3 to 38 years). RESULTS Compared with ε33, age- and sex-adjusted lipoprotein(a) concentrations were lower by 15% in ε23, by 24% in ε24, and by 36% in ε22; adjusted for plasma apolipoprotein E, corresponding values were 22%, 28%, and 62%. These reductions were independent of LPA genotypes. Compared with ε2 carriers with lipoprotein(a) ≤50 mg/dL, the hazard ratio for myocardial infarction was 1.26 (95% confidence interval: 1.06 to 1.49) for ε2 noncarriers with lipoprotein(a) ≤50 mg/dL, 1.68 (1.21 to 2.32) for ε2 carriers with lipoprotein(a) >50 mg/dL, and 1.92 (1.59 to 2.32) for ε2 noncarriers with lipoprotein(a) >50 mg/dL (interaction, P = 0.57); corresponding values for aortic valve stenosis were 1.05 (0.74 to 1.51), 1.49 (0.72 to 3.08), and 2.04 (1.46 to 2.26) (interaction, P = 0.50). Further adjustment for APOE ε2/3/4 genotype had minimal influence on these risk estimates. CONCLUSIONS APOE ε2 is a strong genetic determinant of low lipoprotein(a) concentrations but does not modify the causal association of lipoprotein(a) with myocardial infarction or aortic valve stenosis.
Collapse
Affiliation(s)
- Leonard Kritharides
- Department of Cardiology, Concord Repatriation General Hospital, University of Sydney, Sydney, New South Wales 2139, Australia
- ANZAC Research Institute, University of Sydney, Sydney, New South Wales 2139, Australia
- Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Børge G Nordestgaard
- Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Anne Tybjærg-Hansen
- Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
- Department of Clinical Biochemistry, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Pia R Kamstrup
- Copenhagen General Population Study, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| | - Shoaib Afzal
- Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
- Copenhagen General Population Study, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, 2730 Herlev, Denmark
| |
Collapse
|
31
|
Zewinger S, Kleber ME, Tragante V, McCubrey RO, Schmidt AF, Direk K, Laufs U, Werner C, Koenig W, Rothenbacher D, Mons U, Breitling LP, Brenner H, Jennings RT, Petrakis I, Triem S, Klug M, Filips A, Blankenberg S, Waldeyer C, Sinning C, Schnabel RB, Lackner KJ, Vlachopoulou E, Nygård O, Svingen GFT, Pedersen ER, Tell GS, Sinisalo J, Nieminen MS, Laaksonen R, Trompet S, Smit RAJ, Sattar N, Jukema JW, Groesdonk HV, Delgado G, Stojakovic T, Pilbrow AP, Cameron VA, Richards AM, Doughty RN, Gong Y, Cooper-DeHoff R, Johnson J, Scholz M, Beutner F, Thiery J, Smith JG, Vilmundarson RO, McPherson R, Stewart AFR, Cresci S, Lenzini PA, Spertus JA, Olivieri O, Girelli D, Martinelli NI, Leiherer A, Saely CH, Drexel H, Mündlein A, Braund PS, Nelson CP, Samani NJ, Kofink D, Hoefer IE, Pasterkamp G, Quyyumi AA, Ko YA, Hartiala JA, Allayee H, Tang WHW, Hazen SL, Eriksson N, Held C, Hagström E, Wallentin L, Åkerblom A, Siegbahn A, Karp I, Labos C, Pilote L, Engert JC, Brophy JM, Thanassoulis G, Bogaty P, Szczeklik W, Kaczor M, Sanak M, Virani SS, Ballantyne CM, Lee VV, Boerwinkle E, Holmes MV, Horne BD, Hingorani A, Asselbergs FW, Patel RS, Krämer BK, et alZewinger S, Kleber ME, Tragante V, McCubrey RO, Schmidt AF, Direk K, Laufs U, Werner C, Koenig W, Rothenbacher D, Mons U, Breitling LP, Brenner H, Jennings RT, Petrakis I, Triem S, Klug M, Filips A, Blankenberg S, Waldeyer C, Sinning C, Schnabel RB, Lackner KJ, Vlachopoulou E, Nygård O, Svingen GFT, Pedersen ER, Tell GS, Sinisalo J, Nieminen MS, Laaksonen R, Trompet S, Smit RAJ, Sattar N, Jukema JW, Groesdonk HV, Delgado G, Stojakovic T, Pilbrow AP, Cameron VA, Richards AM, Doughty RN, Gong Y, Cooper-DeHoff R, Johnson J, Scholz M, Beutner F, Thiery J, Smith JG, Vilmundarson RO, McPherson R, Stewart AFR, Cresci S, Lenzini PA, Spertus JA, Olivieri O, Girelli D, Martinelli NI, Leiherer A, Saely CH, Drexel H, Mündlein A, Braund PS, Nelson CP, Samani NJ, Kofink D, Hoefer IE, Pasterkamp G, Quyyumi AA, Ko YA, Hartiala JA, Allayee H, Tang WHW, Hazen SL, Eriksson N, Held C, Hagström E, Wallentin L, Åkerblom A, Siegbahn A, Karp I, Labos C, Pilote L, Engert JC, Brophy JM, Thanassoulis G, Bogaty P, Szczeklik W, Kaczor M, Sanak M, Virani SS, Ballantyne CM, Lee VV, Boerwinkle E, Holmes MV, Horne BD, Hingorani A, Asselbergs FW, Patel RS, Krämer BK, Scharnagl H, Fliser D, März W, Speer T. Relations between lipoprotein(a) concentrations, LPA genetic variants, and the risk of mortality in patients with established coronary heart disease: a molecular and genetic association study. Lancet Diabetes Endocrinol 2017; 5:534-543. [PMID: 28566218 PMCID: PMC5651679 DOI: 10.1016/s2213-8587(17)30096-7] [Show More Authors] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 01/25/2017] [Accepted: 02/14/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Lipoprotein(a) concentrations in plasma are associated with cardiovascular risk in the general population. Whether lipoprotein(a) concentrations or LPA genetic variants predict long-term mortality in patients with established coronary heart disease remains less clear. METHODS We obtained data from 3313 patients with established coronary heart disease in the Ludwigshafen Risk and Cardiovascular Health (LURIC) study. We tested associations of tertiles of lipoprotein(a) concentration in plasma and two LPA single-nucleotide polymorphisms ([SNPs] rs10455872 and rs3798220) with all-cause mortality and cardiovascular mortality by Cox regression analysis and with severity of disease by generalised linear modelling, with and without adjustment for age, sex, diabetes diagnosis, systolic blood pressure, BMI, smoking status, estimated glomerular filtration rate, LDL-cholesterol concentration, and use of lipid-lowering therapy. Results for plasma lipoprotein(a) concentrations were validated in five independent studies involving 10 195 patients with established coronary heart disease. Results for genetic associations were replicated through large-scale collaborative analysis in the GENIUS-CHD consortium, comprising 106 353 patients with established coronary heart disease and 19 332 deaths in 22 studies or cohorts. FINDINGS The median follow-up was 9·9 years. Increased severity of coronary heart disease was associated with lipoprotein(a) concentrations in plasma in the highest tertile (adjusted hazard radio [HR] 1·44, 95% CI 1·14-1·83) and the presence of either LPA SNP (1·88, 1·40-2·53). No associations were found in LURIC with all-cause mortality (highest tertile of lipoprotein(a) concentration in plasma 0·95, 0·81-1·11 and either LPA SNP 1·10, 0·92-1·31) or cardiovascular mortality (0·99, 0·81-1·2 and 1·13, 0·90-1·40, respectively) or in the validation studies. INTERPRETATION In patients with prevalent coronary heart disease, lipoprotein(a) concentrations and genetic variants showed no associations with mortality. We conclude that these variables are not useful risk factors to measure to predict progression to death after coronary heart disease is established. FUNDING Seventh Framework Programme for Research and Technical Development (AtheroRemo and RiskyCAD), INTERREG IV Oberrhein Programme, Deutsche Nierenstiftung, Else-Kroener Fresenius Foundation, Deutsche Stiftung für Herzforschung, Deutsche Forschungsgemeinschaft, Saarland University, German Federal Ministry of Education and Research, Willy Robert Pitzer Foundation, and Waldburg-Zeil Clinics Isny.
Collapse
Affiliation(s)
- Stephen Zewinger
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Marcus E Kleber
- Fifth Department of Medicine, University Heidelberg, Mannheim, Germany; Institute of Nutrition, Friedrich-Schiller University, Jena, Germany
| | - Vinicius Tragante
- Department of Cardiology, Heart and Lungs Division, UMC Utrecht, Utrecht, Netherlands
| | - Raymond O McCubrey
- Intermountain Heart Institute, Intermountain Medical Center, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Amand F Schmidt
- Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK
| | - Kenan Direk
- Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK
| | - Ulrich Laufs
- Department of Internal Medicine III, Saarland University Hospital, Homburg/Saar, Germany
| | - Christian Werner
- Department of Internal Medicine III, Saarland University Hospital, Homburg/Saar, Germany
| | - Wolfgang Koenig
- Department of Internal Medicine II-Cardiology, University of Ulm Medical Centre, Ulm, Germany; Deutsches Herzzentrum München, Technische Universität München, Munich, Germany; German Centre of Cardiovascular Research (DZHK), Partner site Munich Heart Alliance, Munich, Germany
| | - Dietrich Rothenbacher
- Division of Clinical Epidemiology and Ageing Research, German Cancer Centre (DKFZ), Heidelberg, Germany; Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Ute Mons
- Division of Clinical Epidemiology and Ageing Research, German Cancer Centre (DKFZ), Heidelberg, Germany
| | - Lutz P Breitling
- Division of Clinical Epidemiology and Ageing Research, German Cancer Centre (DKFZ), Heidelberg, Germany
| | - Herrmann Brenner
- Network Ageing Research, University Heidelberg, Mannheim, Germany; Division of Clinical Epidemiology and Ageing Research, German Cancer Centre (DKFZ), Heidelberg, Germany
| | - Richard T Jennings
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Ioannis Petrakis
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Sarah Triem
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Mira Klug
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Alexandra Filips
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Stefan Blankenberg
- University Heart Centre Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Christoph Waldeyer
- University Heart Centre Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Christoph Sinning
- University Heart Centre Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Renate B Schnabel
- University Heart Centre Hamburg, Clinic for General and Interventional Cardiology, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK e.V.), partner site Hamburg/Kiel/Lübeck, Germany
| | - Karl J Lackner
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Mainz, Germany
| | | | - Ottar Nygård
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | | | | | - Grethe S Tell
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
| | - Juha Sinisalo
- Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | - Markku S Nieminen
- Heart and Lung Centre, Helsinki University Hospital, Helsinki, Finland
| | - Reijo Laaksonen
- Medical School, Tampere University, Tampere, Finland; Finnish Clinical Biobank Tampere, University Hospital of Tampere, Tampere, Finland
| | - Stella Trompet
- Department of Geriatics and Gerontology, Leiden University Medical Centre, Leiden, Netherlands; Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Roelof A J Smit
- Department of Geriatics and Gerontology, Leiden University Medical Centre, Leiden, Netherlands; Department of Cardiology, Leiden University Medical Centre, Leiden, Netherlands
| | - Naveed Sattar
- Institute of Cardiovascular and Medical Science, BHF Glasgow, Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - J Wouter Jukema
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Centre, Leiden, Netherlands; Interuniversity Cardiology Institute of the Netherlands, Utrecht, Netherlands
| | - Heinrich V Groesdonk
- Department of Anesthesiology, Intensive Care Medicine, and Pain Medicine, Saarland University Hospital, Homburg/Saar, Germany
| | - Graciela Delgado
- Fifth Department of Medicine, University Heidelberg, Mannheim, Germany
| | - Tatjana Stojakovic
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
| | - Anna P Pilbrow
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - Vicky A Cameron
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| | - A Mark Richards
- Christchurch Heart Institute, University of Otago, Christchurch, New Zealand; Cardiovascular Research Institute, National University of Singapore, Singapore
| | - Robert N Doughty
- Heart Health Research Group, University of Auckland, New Zealand
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, Colleges of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Rhonda Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, Colleges of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Julie Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, Colleges of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany; LIFE Research Centre for Civilisation Diseases, University of Leipzig, Leipzig, Germany
| | | | - Joachim Thiery
- LIFE Research Centre for Civilisation Diseases, University of Leipzig, Leipzig, Germany; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital, Leipzig, Germany
| | - J Gustav Smith
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden; Skåne University Hospital, Lund, Sweden
| | - Ragnar O Vilmundarson
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Ruth McPherson
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Alexandre F R Stewart
- Ruddy Canadian Cardiovascular Genetics Centre, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Sharon Cresci
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA; Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
| | - Petra A Lenzini
- Statistical Genomics Division, Department of Genetics, Washington University School of Medicine, Saint Louis, MO, USA
| | - John A Spertus
- Saint Luke's Mid America Heart Institute, Kansas City, MO, USA; Department of Biomedical and Health Informatics, University of Missouri-Kansas City, Kansas City, MO, USA
| | | | | | | | - Andreas Leiherer
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Christoph H Saely
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein
| | - Heinz Drexel
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria; Private University of the Principality of Liechtenstein, Triesen, Liechtenstein; Department of Medicine and Cardiology, Academic Teaching Hospital Feldkirch, Feldkirch, Austria; Drexel University College of Medicine, Philadelphia, PA, USA
| | - Axel Mündlein
- Vorarlberg Institute for Vascular Investigation and Treatment (VIVIT), Feldkirch, Austria
| | - Peter S Braund
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK; Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, UK
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK; Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, BHF Cardiovascular Research Centre, Glenfield Hospital, Leicester, UK; Leicester NIHR Biomedical Research Unit in Cardiovascular Disease, Glenfield Hospital, Leicester, UK
| | - Daniel Kofink
- Department of Cardiology, Heart and Lungs Division, UMC Utrecht, Utrecht, Netherlands
| | - Imo E Hoefer
- Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht, Netherlands
| | - Gerard Pasterkamp
- Laboratory of Experimental Cardiology, UMC Utrecht, Utrecht, Netherlands
| | - Arshed A Quyyumi
- Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - Yi-An Ko
- Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | - Niclas Eriksson
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Claes Held
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Emil Hagström
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Lars Wallentin
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Axel Åkerblom
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden
| | - Agneta Siegbahn
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden; Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Igor Karp
- University of Montreal Hospital Research Centre (CRCHUM), University of Montreal, Montreal, QC, Canada; Department of Social and Preventive Medicine, University of Montreal, Montreal, QC, Canada; Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | | | - Louise Pilote
- Department of Medicine, McGill University, Montreal, QC, Canada; Division of General Internal Medicine, McGill University Health Centre, Montreal, QC, Canada; Division of Clinical Epidemiology, McGill University Health Centre, Montreal, QC, Canada
| | - James C Engert
- Department of Medicine, McGill University, Montreal, QC, Canada
| | - James M Brophy
- Department of Medicine, McGill University, Montreal, QC, Canada
| | | | - Peter Bogaty
- Department of Medicine, Université Laval, QC, Canada
| | | | - Marcin Kaczor
- Jagielonian University Medical College, Kraków, Poland
| | - Marek Sanak
- Jagielonian University Medical College, Kraków, Poland
| | - Salim S Virani
- Section of Cardiology, Michael E DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, TX, USA
| | - Christie M Ballantyne
- Section of Cardiovascular Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Vei-Vei Lee
- Department of Biostatistics 7, Epidemiology, Texas Heart Institute, Houston, TX, USA
| | - Eric Boerwinkle
- School of Public Health, University of Texas, Houston, TX, USA
| | - Michael V Holmes
- Medical Research Council Population Health Research Unit at the University of Oxford, University of Oxford, Oxford, UK; Clinical Trial Service Unit and Epidemiological Studies Unit (CTSU), Nuffield Department of Population Health, University of Oxford, Oxford, UK; National Institute for Health Research Oxford Biomedical Research Centre, Oxford University Hospital, Oxford, UK
| | - Benjamin D Horne
- Intermountain Heart Institute, Intermountain Medical Center, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Aroon Hingorani
- Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK
| | - Folkert W Asselbergs
- Department of Cardiology, Heart and Lungs Division, UMC Utrecht, Utrecht, Netherlands; Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK; Durrer Centre of Cardiogenetic Research, ICIN-Netherlands Heart Institute, Utrecht, Netherlands
| | - Riyaz S Patel
- Institute of Cardiovascular Science Facultyof Population Health Science, University College London, London, UK
| | | | - Bernhard K Krämer
- Fifth Department of Medicine, University Heidelberg, Mannheim, Germany
| | - Hubert Scharnagl
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria
| | - Danilo Fliser
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| | - Winfried März
- Fifth Department of Medicine, University Heidelberg, Mannheim, Germany; Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University Graz, Graz, Austria; Synlab Academy, Synlab Holding, Mannheim, Germany.
| | - Thimoteus Speer
- Department of Internal Medicine IV, Saarland University Hospital, Homburg/Saar, Germany
| |
Collapse
|
32
|
Mack S, Coassin S, Rueedi R, Yousri NA, Seppälä I, Gieger C, Schönherr S, Forer L, Erhart G, Marques-Vidal P, Ried JS, Waeber G, Bergmann S, Dähnhardt D, Stöckl A, Raitakari OT, Kähönen M, Peters A, Meitinger T, Strauch K, Kedenko L, Paulweber B, Lehtimäki T, Hunt SC, Vollenweider P, Lamina C, Kronenberg F. A genome-wide association meta-analysis on lipoprotein (a) concentrations adjusted for apolipoprotein (a) isoforms. J Lipid Res 2017; 58:1834-1844. [PMID: 28512139 PMCID: PMC5580897 DOI: 10.1194/jlr.m076232] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/04/2017] [Indexed: 12/13/2022] Open
Abstract
High lipoprotein (a) [Lp(a)] concentrations are an independent risk factor for cardiovascular outcomes. Concentrations are strongly influenced by apo(a) kringle IV repeat isoforms. We aimed to identify genetic loci associated with Lp(a) concentrations using data from five genome-wide association studies (n = 13,781). We identified 48 independent SNPs in the LPA and 1 SNP in the APOE gene region to be significantly associated with Lp(a) concentrations. We also adjusted for apo(a) isoforms to identify loci affecting Lp(a) levels independently from them, which resulted in 31 SNPs (30 in the LPA, 1 in the APOE gene region). Seven SNPs showed a genome-wide significant association with coronary artery disease (CAD) risk. A rare SNP (rs186696265; MAF ∼1%) showed the highest effect on Lp(a) and was also associated with increased risk of CAD (odds ratio = 1.73, P = 3.35 × 10−30). Median Lp(a) values increased from 2.1 to 91.1 mg/dl with increasing number of Lp(a)-increasing alleles. We found the APOE2-determining allele of rs7412 to be significantly associated with Lp(a) concentrations (P = 3.47 × 10−10). Each APOE2 allele decreased Lp(a) by 3.34 mg/dl corresponding to ∼15% of the population’s mean values. Performing a gene-based test of association, including suspected Lp(a) receptors and regulators, resulted in one significant association of the TLR2 gene with Lp(a) (P = 3.4 × 10−4). In summary, we identified a large number of independent SNPs in the LPA gene region, as well as the APOE2 allele, to be significantly associated with Lp(a) concentrations.
Collapse
Affiliation(s)
- Salome Mack
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Stefan Coassin
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Rico Rueedi
- Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland.,Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Noha A Yousri
- Department of Physiology and Biophysics, Weill Cornell Medical College-Qatar, Doha, Qatar.,Department of Computer and Systems Engineering, Alexandria University, 21526 Alexandria, Egypt
| | - Ilkka Seppälä
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, 33520 Tampere, Finland
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany.,Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany.,Research Unit of Molecular Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Sebastian Schönherr
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Lukas Forer
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Gertraud Erhart
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Pedro Marques-Vidal
- Department of Medicine, Internal Medicine, Lausanne University Hospital, 1015 Lausanne, Switzerland
| | - Janina S Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Gerard Waeber
- Department of Medicine, Internal Medicine, Lausanne University Hospital, 1015 Lausanne, Switzerland
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland.,Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Doreen Dähnhardt
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Andrea Stöckl
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Olli T Raitakari
- Department of Clinical Physiology, Turku University Hospital, 20520 Turku, Finland.,Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, 20520 Turku, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital and University of Tampere, 33521 Tampere, Finland
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany.,German Centre for Cardiovascular Research (DZHK), 80802 Munich, Germany.,German Center for Diabetes Research (DZD e.V.), 85764 Neuherberg, Germany
| | - Thomas Meitinger
- Institute of Human Genetics, Technische Universität München, 81675 München, Germany.,Institute of Human Genetics, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany.,Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany.,Institute of Medical Informatics, Biometry, and Epidemiology, Ludwig-Maximilians-Universität, 81377 Munich, Germany
| | | | - Ludmilla Kedenko
- First Department of Internal Medicine, Paracelsus Private Medical University, 5020 Salzburg, Austria
| | - Bernhard Paulweber
- First Department of Internal Medicine, Paracelsus Private Medical University, 5020 Salzburg, Austria
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories and University of Tampere School of Medicine, 33520 Tampere, Finland
| | - Steven C Hunt
- Cardiovascular Genetics Division, University of Utah School of Medicine, Salt Lake City, UT 84108.,Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Peter Vollenweider
- Department of Medicine, Internal Medicine, Lausanne University Hospital, 1015 Lausanne, Switzerland
| | - Claudia Lamina
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Florian Kronenberg
- Division of Genetic Epidemiology, Department of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
33
|
Are genetic polymorphisms in the renin-angiotensin-aldosterone system associated with essential hypertension? Evidence from genome-wide association studies. J Hum Hypertens 2017; 31:695-698. [PMID: 28425437 DOI: 10.1038/jhh.2017.29] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/20/2017] [Accepted: 03/03/2017] [Indexed: 01/01/2023]
Abstract
In candidate gene era, dozens of single-nucleotide polymorphisms (SNPs) within renin-angiotensin-aldosterone system (RAAS) have been reported to be significantly associated with hypertension. However, the unbiased genome-wide association studies (GWAS) rarely identified the SNPs within RAAS were associated with hypertension or blood pressure (BP) traits. In order to figure out whether genetic polymorphisms of RAAS are really associated with hypertension, we systemically searched the GWAS Catalogue and identified all the known RAAS genes and relevant diseases/traits. After data processing, we found that polymorphisms within REN, AGT, ACE2, CYP11B2, ATP6AP2 and HSD11B2 were not associated with any disease or trait. SNPs within ACE, AGTR1, AGTR2, MAS1, RENBP and NR3C2 were associated with other diseases or traits, but showed no direct connection with hypertension. The only SNP associated with a BP trait, systolic BP was rs17367504. However, it is located in the intronic region of MTHFR near many plausible candidate genes, including CLCN6, NPPA, NPPB and AGTRAP. Therefore, the effect of RAAS polymorphisms may have been overestimated during the 'candidate gene era'. In the time of 'precision medicine', the power of RAAS variants needs to be reconsidered when evaluating one's susceptibility of hypertension.
Collapse
|
34
|
Abstract
Lipoprotein(a) [Lp(a)] is a highly atherogenic lipoprotein that is under strong genetic control by the LPA gene locus. Genetic variants including a highly polymorphic copy number variation of the so called kringle IV repeats at this locus have a pronounced influence on Lp(a) concentrations. High concentrations of Lp(a) as well as genetic variants which are associated with high Lp(a) concentrations are both associated with cardiovascular disease which very strongly supports causality between Lp(a) concetrations and cardiovascular disease. This method of using a genetic variant that has a pronounced influence on a biomarker to support causality with an outcome is called Mendelian randomization approach and was applied for the first time two decades ago with data from Lp(a) and cardiovascular disease. This approach was also used to demonstrate a causal association between high Lp(a) concentrations and aortic valve stenosis, between low concentrations and type-2 diabetes mellitus and to exclude a causal association between Lp(a) concentrations and venous thrombosis. Considering the high frequency of these genetic variants in the population makes Lp(a) the strongest genetic risk factor for cardiovascular disease identified so far. Promising drugs that lower Lp(a) are on the horizon but their efficacy in terms of reducing clinical outcomes still has to be shown.
Collapse
|
35
|
Schmidt K, Noureen A, Kronenberg F, Utermann G. Structure, function, and genetics of lipoprotein (a). J Lipid Res 2016; 57:1339-59. [PMID: 27074913 DOI: 10.1194/jlr.r067314] [Citation(s) in RCA: 373] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Indexed: 12/29/2022] Open
Abstract
Lipoprotein (a) [Lp(a)] has attracted the interest of researchers and physicians due to its intriguing properties, including an intragenic multiallelic copy number variation in the LPA gene and the strong association with coronary heart disease (CHD). This review summarizes present knowledge of the structure, function, and genetics of Lp(a) with emphasis on the molecular and population genetics of the Lp(a)/LPA trait, as well as aspects of genetic epidemiology. It highlights the role of genetics in establishing Lp(a) as a risk factor for CHD, but also discusses uncertainties, controversies, and lack of knowledge on several aspects of the genetic Lp(a) trait, not least its function.
Collapse
Affiliation(s)
- Konrad Schmidt
- Divisions of Human Genetics Medical University of Innsbruck, Innsbruck, Austria Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Asma Noureen
- Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Kronenberg
- Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerd Utermann
- Divisions of Human Genetics Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|