1
|
Wheeler AM, Riley TR, Merriman TR. Genetic Risk Scores for the Clinical Rheumatologist. J Clin Rheumatol 2025; 31:26-32. [PMID: 39454094 DOI: 10.1097/rhu.0000000000002152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
BACKGROUND/HISTORICAL PERSPECTIVE The advent of genome-wide sequencing and large-scale genetic epidemiological studies has led to numerous opportunities for the application of genetics in clinical medicine. Leveraging this information toward the formation of clinically useful tools has been an ongoing research goal in this area. A genetic risk score (GRS) is a measure that attempts to estimate the cumulative contribution of established genetic risk factors toward an outcome of interest, taking into account the cumulative risk that each of these individual genetic risk factors conveys. The purpose of this perspective is to provide a systematic framework to evaluate a GRS for clinical application. SUMMARY OF CURRENT LITERATURE Since the initial polygenic risk score methodology in 2007, there has been increasing GRS application across the medical literature. In rheumatology, this has included application to rheumatoid arthritis, gout, spondyloarthritis, lupus, and inflammatory arthritis. MAJOR CONCLUSIONS GRSs are particularly relevant to rheumatology, where common diseases have many complex genetic factors contributing to risk. Despite this, there is no widely accepted method for the critical application of a GRS, which can be a particular challenge for the clinical rheumatologist seeking to clinically apply GRSs. This review provides a framework by which the clinician may systematically evaluate a GRS. FUTURE RESEARCH DIRECTIONS As genotyping becomes more accessible and cost-effective, it will become increasingly important to recognize the clinical applicability of GRSs and identify those of the highest utility for patient care. This framework for the evaluation of a GRS will also help ensure reliability among GRS research in rheumatology, thereby helping to advance the field.
Collapse
Affiliation(s)
- Austin M Wheeler
- From the University of Nebraska Medical Center and VA Nebraska-Western Iowa Health Care System, Omaha, NE
| | - Thomas R Riley
- University of Pennsylvania and Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | | |
Collapse
|
2
|
Sun C, Cheng X, Xu J, Chen H, Tao J, Dong Y, Wei S, Chen R, Meng X, Ma Y, Tian H, Guo X, Bi S, Zhang C, Kang J, Zhang M, Lv H, Shang Z, Lv W, Zhang R, Jiang Y. A review of disease risk prediction methods and applications in the omics era. Proteomics 2024; 24:e2300359. [PMID: 38522029 DOI: 10.1002/pmic.202300359] [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: 09/15/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
Risk prediction and disease prevention are the innovative care challenges of the 21st century. Apart from freeing the individual from the pain of disease, it will lead to low medical costs for society. Until very recently, risk assessments have ushered in a new era with the emergence of omics technologies, including genomics, transcriptomics, epigenomics, proteomics, and so on, which potentially advance the ability of biomarkers to aid prediction models. While risk prediction has achieved great success, there are still some challenges and limitations. We reviewed the general process of omics-based disease risk model construction and the applications in four typical diseases. Meanwhile, we highlighted the problems in current studies and explored the potential opportunities and challenges for future clinical practice.
Collapse
Affiliation(s)
- Chen Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Xiangshu Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Jing Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Haiyan Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Junxian Tao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Yu Dong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Siyu Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Rui Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xin Meng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yingnan Ma
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| | - Hongsheng Tian
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xuying Guo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shuo Bi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Chen Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Jingxuan Kang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Mingming Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zhenwei Shang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Wenhua Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Ruijie Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The EWAS Project, Harbin, China
| |
Collapse
|
3
|
Matthews LJ, Zhang Z, Martschenko DO. Schoolhouse risk: Can we mitigate the polygenic Pygmalion effect? Acta Psychol (Amst) 2024; 248:104403. [PMID: 39003994 PMCID: PMC11343671 DOI: 10.1016/j.actpsy.2024.104403] [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: 12/15/2023] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND Although limited in predictive accuracy, polygenic scores (PGS) for educational outcomes are currently available to the public via direct-to-consumer genetic testing companies. Further, there is a growing movement to apply PGS in educational settings via 'precision education.' Prior scholarship highlights the potentially negative impacts of such applications, as disappointing results may give rise a "polygenic Pygmalion effect." In this paper two studies were conducted to identify factors that may mitigate or exacerbate negative impacts of PGS. METHODS Two studies were conducted. In each, 1188 students were randomized to one of four conditions: Low-percentile polygenic score for educational attainment (EA-PGS), Low EA-PGS + Mitigating information, Low EA-PGS + Exacerbating information, or Control. Regression analyses were used to examine differences between conditions. RESULTS In Study 1, participants randomized to Control reported significantly higher on the Rosenberg Self-Esteem Scale (RSES), Competence Scale (CS), Academic Efficacy Scale (AES) and Educational Potential Scale (EPS). CS was significantly higher in the Low EA-PGS + Mitigating information condition. CS and AES were significantly lower in the Low EA-PGS + Exacerbating information condition compared to the Low EA-PGS + Mitigating information condition. In Study 2, participants randomized to Control reported significantly higher CS and AES. Pairwise comparisons did not show significant differences in CS and AES. Follow-up pairwise comparisons using Tukey P-value correction did not find significant associations between non-control conditions. CONCLUSION These studies replicated the polygenic Pygmalion effect yet were insufficiently powered to detect significant effects of mitigating contextual information. Regardless of contextual information, disappointing EA-PGS results were significantly associated with lower assessments of self-esteem, competence, academic efficacy, and educational potential.
Collapse
Affiliation(s)
- Lucas J Matthews
- Columbia University, Department of Medical Humanities & Ethics, New York, NY, United States; The Hastings Center, New York, NY, United States.
| | - Zhijun Zhang
- New York State Psychiatric Institute, Department of Mental Health and Data Science, New York, NY, United States.
| | - Daphne O Martschenko
- Stanford Center for Biomedical Ethics and Department of Pediatrics, Stanford University; Stanford, CA, United States.
| |
Collapse
|
4
|
Rodríguez-Gutiérrez PG, Hernández-Flores TDJ, Zepeda-Olmos PM, Reyes-Rodríguez CD, Robles-Espinoza K, Solís-Gómez U, González-García JR, Magaña-Torres MT. High Prevalence of Familial Hypercholesterolemia Due to the Founder Effect of the LDLR c.2271del Variant in Communities of Oaxaca, Mexico. Arch Med Res 2024; 55:102971. [PMID: 38513336 DOI: 10.1016/j.arcmed.2024.102971] [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: 08/11/2023] [Revised: 01/31/2024] [Accepted: 02/13/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION In Mexico, familial hypercholesterolemia (FH) is underdiagnosed, but population screening in small communities where at least one homozygous patient has already been detected results in a useful and inexpensive approach to reduce this problem. Considering that we previously reported nine homozygous cases from the state of Oaxaca, we decided to perform a population screening to identify patients with FH and to describe both their biochemical and genetic characteristics. METHODS LDL cholesterol (LDLc) was quantified in 2,093 individuals from 11 communities in Oaxaca; either adults with LDLc levels ≥170 mg/dL or children with LDLc ≥130 mg/dL were classified as suggestive of FH and therefore included in the genetic study. LDLR and APOB (547bp fragment of exon 26) genes were screened by sequencing and MLPA analysis. RESULTS Two hundred and five individuals had suggestive FH, with a mean LDLc of 223 ± 54 mg/dL (range: 131-383 mg/dL). Two pathogenic variants in the LDLR gene were detected in 149 individuals: c.-139_-130del (n = 1) and c.2271del (n = 148). All patients had a heterozygous genotype. With the cascade screening of their relatives (n = 177), 15 heterozygous individuals for the c.2271del variant were identified, presenting a mean LDLc of 133 ± 35 mg/dL (range: 60-168 mg/dL). CONCLUSIONS The FH frequency in this study was 7.8% (164/2093), the highest reported worldwide. A founder effect combined with inbreeding could be responsible for the high percentage of patients with the LDLR c.2271del variant (99.4%), which allowed us to detect both significant biochemical heterogeneity and incomplete penetrance; hence, we assumed the presence of phenotype-modifying variants.
Collapse
Affiliation(s)
- Perla Graciela Rodríguez-Gutiérrez
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México; División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Teresita de Jesús Hernández-Flores
- Departamento de Disciplinas Filosófico, Metodológicas e Instrumentales. Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Paola Montserrat Zepeda-Olmos
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México; División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Christian Daniel Reyes-Rodríguez
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México; División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Kiabeth Robles-Espinoza
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, Jalisco, México; División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - Ulises Solís-Gómez
- Hospital Regional, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Tepic Aquiles Calles Ramírez, Tepic, Nayarit, México
| | - Juan Ramón González-García
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México
| | - María Teresa Magaña-Torres
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, Jalisco, México.
| |
Collapse
|
5
|
Paquette M, Baass A. Advances in familial hypercholesterolemia. Adv Clin Chem 2024; 119:167-201. [PMID: 38514210 DOI: 10.1016/bs.acc.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Familial hypercholesterolemia (FH), a semi-dominant genetic disease affecting more than 25 million people worldwide, is associated with severe hypercholesterolemia and premature atherosclerotic cardiovascular disease. Over the last decade, advances in data analysis, screening, diagnosis and cardiovascular risk stratification has significantly improved our ability to deliver precision medicine for these patients. Furthermore, recent updates on guideline recommendations and new therapeutic approaches have also proven to be highly beneficial. It is anticipated that both ongoing and upcoming clinical trials will offer further insights for the care and treatment of FH patients.
Collapse
Affiliation(s)
- Martine Paquette
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal, QC, Canada
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Montreal, QC, Canada; Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
6
|
Sawhney JPS, Madan K. Familial hypercholesterolemia. Indian Heart J 2024; 76 Suppl 1:S108-S112. [PMID: 38599725 PMCID: PMC11019323 DOI: 10.1016/j.ihj.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/02/2023] [Indexed: 04/12/2024] Open
Abstract
Familial hypercholesterolemia is a common genetic disorder of autosomal inheritance associated with elevated LDL-cholesterol. It is estimated to affect 1:250 individuals in general population roughly estimated to be 5 million in India. The prevalence of FH is higher in young CAD patients (<55 years in men; <60 years in women). FH is underdiagnosed and undertreated. Screening during childhood and Cascade screening of family members of known FH patients is of utmost importance in order to prevent the burden of CAD. Early identification of FH patients and early initiation of the lifelong lipid lowering therapy is the most effective strategy for managing FH. FH management includes pharmaceutical agents (statins and non statin drugs) and lifestyle modification. Inspite of maximum dose of statin with or without Ezetimibe, if target levels of LDL-C are not achieved, Bempedoic acid, proprotein convertase subtilisin/kexin type 9 (PCSK9) Inhibitors/Inclisiran can be added.
Collapse
Affiliation(s)
- J P S Sawhney
- Dharma Vira Heart Center, Sir Ganga Ram Hospital, New Delhi 110060, India.
| | - Kushal Madan
- Dharma Vira Heart Center, Sir Ganga Ram Hospital, New Delhi 110060, India.
| |
Collapse
|
7
|
Gupta R. Genetics-based risk scores for prediction of premature coronary artery disease. Indian Heart J 2023; 75:327-334. [PMID: 37633460 PMCID: PMC10568063 DOI: 10.1016/j.ihj.2023.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 07/24/2023] [Accepted: 08/20/2023] [Indexed: 08/28/2023] Open
Abstract
Premature coronary artery disease (CAD) is endemic in India. Global Burden of Diseases study has reported that it led to 286,000 deaths in 2019 in India. Many of these patients have standard risk factors but a third have none. Clinical risk algorithms and imaging provide limited risk information in premature CAD. CAD is multifactorial and studies have now focused on the predictive capability of clusters of genes and single nucleotide polymorphisms (SNPs) using gene risk score (GRS). Older studies combined data from 10 to 12 genes and 100-500 SNPs to calculate GRS, however, following the advent of genome-wide association studies (GWAS), millions of SNPs have been incorporated. Studies have reported that GWAS-based GRS may be more discriminative than conventional tools. Recent studies, especially among South Asians, have reported that GRS improves net reclassification by 15% (12-19%) for younger individuals. Aggressive lifestyle interventions and lipid-lowering therapies can ameliorate risk in high-GRS individuals and potentially prevent premature CAD.
Collapse
Affiliation(s)
- Rajeev Gupta
- Department of Preventive Cardiology & Medicine, Eternal Heart Care Centre & Research Institute, Jaipur, India.
| |
Collapse
|
8
|
Noda K, Hattori Y, Hori M, Nakaoku Y, Tanaka A, Yoshimoto T, Nishimura K, Yokota T, Harada-Shiba M, Ihara M. Amplified Risk of Intracranial Artery Stenosis/Occlusion Associated With RNF213 p.R4810K in Familial Hypercholesterolemia. JACC. ASIA 2023; 3:625-633. [PMID: 37614551 PMCID: PMC10442882 DOI: 10.1016/j.jacasi.2023.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/23/2023] [Accepted: 03/18/2023] [Indexed: 08/25/2023]
Abstract
Background The RNF213 p.R4810K variant is associated with moyamoya disease in East Asian individuals and increases the risk of developing intracranial major artery stenosis/occlusion (ICASO) that affects anterior circulation. Meanwhile, 0.5% to 2.5% of asymptomatic East Asian individuals also carry this variant. As such, additional factors are likely required to develop ICASO in variant carriers. Familial hypercholesterolemia (FH) is a common genetic disorder in Japan that has a significant associated risk of developing premature coronary atherosclerosis; however, the relationship between ICASO and FH remains unknown. Objectives This study aimed to determine if FH facilitates RNF213 p.R4810K carriers to develop ICASO. Methods We enrolled patients with FH who had undergone brain magnetic resonance angiography at our hospital from May 2005 to March 2020. The RNF213 p.R4810K variant, and LDLR and PCSK9 mutations were genotyped. ICASO lesions in the brain magnetic resonance angiogram were analyzed. Results Six RNF213 p.R4810K variant carriers were identified among 167 patients with FH (LDLR, n = 104; PCSK9, n = 22). Five of the carriers (83.3%) exhibited ICASO in the anterior circulation; a significant difference in ICASO frequency was observed between the variant carriers and noncarriers (P = 0.025). The median number of stenotic or occluded arteries in the anterior circulation was also significantly larger in the variant carriers (3 vs 1, P = 0.01); however, did not differ between patients with FH with LDLR and PCSK9 mutations. Conclusions Patients with FH exhibit increased prevalence and severity of ICASO associated with RNF213 p.R4810K. Gene mutations for FH may confer an increased risk of ICASO in RNF213 p.R4810K carriers.
Collapse
Affiliation(s)
- Kotaro Noda
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yorito Hattori
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Mika Hori
- Department of Endocrinology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Yuriko Nakaoku
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Akito Tanaka
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Takeshi Yoshimoto
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Kunihiro Nishimura
- Department of Preventive Medicine and Epidemiology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Takanori Yokota
- Department of Neurology and Neurological Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Mariko Harada-Shiba
- Cardiovascular Center, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| |
Collapse
|
9
|
Lan NSR, Bajaj A, Watts GF, Cuchel M. Recent advances in the management and implementation of care for familial hypercholesterolaemia. Pharmacol Res 2023; 194:106857. [PMID: 37460004 DOI: 10.1016/j.phrs.2023.106857] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/07/2023] [Accepted: 07/14/2023] [Indexed: 07/22/2023]
Abstract
Familial hypercholesterolaemia (FH) is a common autosomal semi-dominant and highly penetrant disorder of the low-density lipoprotein (LDL) receptor pathway, characterised by lifelong elevated levels of low-density lipoprotein cholesterol (LDL-C) and increased risk of atherosclerotic cardiovascular disease (ASCVD). However, many patients with FH are not diagnosed and do not attain recommended LDL-C goals despite maximally tolerated doses of potent statin and ezetimibe. Over the past decade, several cholesterol-lowering therapies such as those targeting proprotein convertase subtilisin/kexin type 9 (PCSK9) or angiopoietin-like 3 (ANGPTL3) with monoclonal antibody or ribonucleic acid (RNA) approaches have been developed that promise to close the treatment gap. The availability of new therapies with complementary modes of action of lipid metabolism has enabled many patients with FH to attain guideline-recommended LDL-C goals. Emerging therapies for FH include liver-directed gene transfer of the LDLR, vaccines targeting key proteins involved in cholesterol metabolism, and CRISPR-based gene editing of PCSK9 and ANGPTL3, but further clinical trials are required. In this review, current and emerging treatment strategies for lowering LDL-C, and ASCVD risk-stratification, as well as implementation strategies for the care of patients with FH are reviewed.
Collapse
Affiliation(s)
- Nick S R Lan
- Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia; School of Medicine, The University of Western Australia, Perth, Australia.
| | - Archna Bajaj
- Division of Translational Medicine & Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gerald F Watts
- Departments of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, Australia; School of Medicine, The University of Western Australia, Perth, Australia
| | - Marina Cuchel
- Division of Translational Medicine & Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Patel AP, Wang M, Ruan Y, Koyama S, Clarke SL, Yang X, Tcheandjieu C, Agrawal S, Fahed AC, Ellinor PT, Tsao PS, Sun YV, Cho K, Wilson PWF, Assimes TL, van Heel DA, Butterworth AS, Aragam KG, Natarajan P, Khera AV. A multi-ancestry polygenic risk score improves risk prediction for coronary artery disease. Nat Med 2023; 29:1793-1803. [PMID: 37414900 PMCID: PMC10353935 DOI: 10.1038/s41591-023-02429-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 05/30/2023] [Indexed: 07/08/2023]
Abstract
Identification of individuals at highest risk of coronary artery disease (CAD)-ideally before onset-remains an important public health need. Prior studies have developed genome-wide polygenic scores to enable risk stratification, reflecting the substantial inherited component to CAD risk. Here we develop a new and significantly improved polygenic score for CAD, termed GPSMult, that incorporates genome-wide association data across five ancestries for CAD (>269,000 cases and >1,178,000 controls) and ten CAD risk factors. GPSMult strongly associated with prevalent CAD (odds ratio per standard deviation 2.14, 95% confidence interval 2.10-2.19, P < 0.001) in UK Biobank participants of European ancestry, identifying 20.0% of the population with 3-fold increased risk and conversely 13.9% with 3-fold decreased risk as compared with those in the middle quintile. GPSMult was also associated with incident CAD events (hazard ratio per standard deviation 1.73, 95% confidence interval 1.70-1.76, P < 0.001), identifying 3% of healthy individuals with risk of future CAD events equivalent to those with existing disease and significantly improving risk discrimination and reclassification. Across multiethnic, external validation datasets inclusive of 33,096, 124,467, 16,433 and 16,874 participants of African, European, Hispanic and South Asian ancestry, respectively, GPSMult demonstrated increased strength of associations across all ancestries and outperformed all available previously published CAD polygenic scores. These data contribute a new GPSMult for CAD to the field and provide a generalizable framework for how large-scale integration of genetic association data for CAD and related traits from diverse populations can meaningfully improve polygenic risk prediction.
Collapse
Affiliation(s)
- Aniruddh P Patel
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Minxian Wang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China.
| | - Yunfeng Ruan
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Satoshi Koyama
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Veteran Affairs Boston Healthcare System, Boston, MA, USA
| | - Shoa L Clarke
- Stanford University School of Medicine, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Xiong Yang
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | | | - Saaket Agrawal
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Akl C Fahed
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Patrick T Ellinor
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Philip S Tsao
- Stanford University School of Medicine, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Yan V Sun
- Veteran Affairs Atlanta Healthcare System, Decatur, GA, USA
| | - Kelly Cho
- Veteran Affairs Boston Healthcare System, Boston, MA, USA
| | | | - Themistocles L Assimes
- Stanford University School of Medicine, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - David A van Heel
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Adam S Butterworth
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, and Centre of Research Excellence, University of Cambridge, Cambridge, UK
| | - Krishna G Aragam
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Pradeep Natarajan
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Amit V Khera
- Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
- Verve Therapeutics, Boston, MA, USA.
| |
Collapse
|
11
|
Li C, Pan Y, Zhang R, Huang Z, Li D, Han Y, Larkin C, Rao V, Sun X, Kelly TN. Genomic Innovation in Early Life Cardiovascular Disease Prevention and Treatment. Circ Res 2023; 132:1628-1647. [PMID: 37289909 PMCID: PMC10328558 DOI: 10.1161/circresaha.123.321999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality globally. Although CVD events do not typically manifest until older adulthood, CVD develops gradually across the life-course, beginning with the elevation of risk factors observed as early as childhood or adolescence and the emergence of subclinical disease that can occur in young adulthood or midlife. Genomic background, which is determined at zygote formation, is among the earliest risk factors for CVD. With major advances in molecular technology, including the emergence of gene-editing techniques, along with deep whole-genome sequencing and high-throughput array-based genotyping, scientists now have the opportunity to not only discover genomic mechanisms underlying CVD but use this knowledge for the life-course prevention and treatment of these conditions. The current review focuses on innovations in the field of genomics and their applications to monogenic and polygenic CVD prevention and treatment. With respect to monogenic CVD, we discuss how the emergence of whole-genome sequencing technology has accelerated the discovery of disease-causing variants, allowing comprehensive screening and early, aggressive CVD mitigation strategies in patients and their families. We further describe advances in gene editing technology, which might soon make possible cures for CVD conditions once thought untreatable. In relation to polygenic CVD, we focus on recent innovations that leverage findings of genome-wide association studies to identify druggable gene targets and develop predictive genomic models of disease, which are already facilitating breakthroughs in the life-course treatment and prevention of CVD. Gaps in current research and future directions of genomics studies are also discussed. In aggregate, we hope to underline the value of leveraging genomics and broader multiomics information for characterizing CVD conditions, work which promises to expand precision approaches for the life-course prevention and treatment of CVD.
Collapse
Affiliation(s)
- Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (C. Li, R.Z., Z.H., X.S.)
| | - Yang Pan
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Ruiyuan Zhang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (C. Li, R.Z., Z.H., X.S.)
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (C. Li, R.Z., Z.H., X.S.)
| | - Davey Li
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Yunan Han
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Claire Larkin
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Varun Rao
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| | - Xiao Sun
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA (C. Li, R.Z., Z.H., X.S.)
| | - Tanika N Kelly
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois Chicago (Y.P., D.L., Y.H., C.L., V.R., T.N.K.)
| |
Collapse
|
12
|
Phulka JS, Ashraf M, Bajwa BK, Pare G, Laksman Z. Current State and Future of Polygenic Risk Scores in Cardiometabolic Disease: A Scoping Review. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2023; 16:286-313. [PMID: 37035923 DOI: 10.1161/circgen.122.003834] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
Abstract
A polygenic risk score (PRS) is derived from a genome-wide association study and represents an aggregate of thousands of single-nucleotide polymorphisms that provide a baseline estimate of an individual's genetic risk for a specific disease or trait at birth. However, it remains unclear how PRSs can be used in clinical practice. We provide an overview of the PRSs related to cardiometabolic disease and discuss the evidence supporting their clinical applications and limitations. The Preferred Reporting Items For Systematic Reviews and Meta-Analysis Extension for Scoping Reviews protocol was used to conduct a scoping review of the MEDLINE, EMBASE, and CENTRAL databases. Across the 4863 studies screened, 82 articles met the inclusion criteria. The most common PRS related to coronary artery disease, followed by hypertension and cerebrovascular disease. Limited ancestral diversity was observed in the study sample populations. Most studies included only individuals of European ancestry. The predictive performance of most PRSs was similar to or superior to traditional risk factors. More than half of the included studies reported an integrated risk model combining a derived PRS and clinical risk tools such as the Framingham Risk Score and Pooled Cohort Equations. The inclusion of a PRS into a clinical risk model tended to improve predictive accuracy consistently. This scoping review is the first of its kind and reports strong evidence for the clinical utility of PRSs in coronary artery disease, hypertension, cerebrovascular disease, and atrial fibrillation. However, most PRSs are generated in cohorts of European ancestry, which likely contributes to a lack of PRS transferability across different ancestral groups. Future prospective studies should focus on further establishing the clinical utility of PRSs and ensuring diversity is incorporated into genome-wide association study cohorts.
Collapse
Affiliation(s)
- Jobanjit S Phulka
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| | - Mishal Ashraf
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| | - Beenu K Bajwa
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| | - Guillaume Pare
- Population Health Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute; Thrombosis and Atherosclerosis Research Institute, Department of Health Research Methods, Evidence, and Impact, Department of Pathology & Molecular Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, ON, Canada (G.P.)
| | - Zachary Laksman
- Heart Rhythm Services & Center for Cardiovascular Innovation, Division of Cardiology, University of British Columbia, Vancouver (J.S.P., M.A., B.K.B., Z.L.)
| |
Collapse
|
13
|
Chiu CH, Hsuan CF, Lin SH, Hung YJ, Hwu CM, Hee SW, Lin SW, Fong SW, Hsieh PCH, Yang WS, Lin WC, Lee HL, Hsieh ML, Li WY, Lin JW, Hsu CN, Wu VC, Chuang GT, Chang YC, Chuang LM. ER ribosomal-binding protein 1 regulates blood pressure and potassium homeostasis by modulating intracellular renin trafficking. J Biomed Sci 2023; 30:13. [PMID: 36803854 PMCID: PMC9940419 DOI: 10.1186/s12929-023-00905-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/07/2023] [Indexed: 02/21/2023] Open
Abstract
BACKGROUND Genome-wide association studies (GWASs) have linked RRBP1 (ribosomal-binding protein 1) genetic variants to atherosclerotic cardiovascular diseases and serum lipoprotein levels. However, how RRBP1 regulates blood pressure is unknown. METHODS To identify genetic variants associated with blood pressure, we performed a genome-wide linkage analysis with regional fine mapping in the Stanford Asia-Pacific Program for Hypertension and Insulin Resistance (SAPPHIRe) cohort. We further investigated the role of the RRBP1 gene using a transgenic mouse model and a human cell model. RESULTS In the SAPPHIRe cohort, we discovered that genetic variants of the RRBP1 gene were associated with blood pressure variation, which was confirmed by other GWASs for blood pressure. Rrbp1- knockout (KO) mice had lower blood pressure and were more likely to die suddenly from severe hyperkalemia caused by phenotypically hyporeninemic hypoaldosteronism than wild-type controls. The survival of Rrbp1-KO mice significantly decreased under high potassium intake due to lethal hyperkalemia-induced arrhythmia and persistent hypoaldosteronism, which could be rescued by fludrocortisone. An immunohistochemical study revealed renin accumulation in the juxtaglomerular cells of Rrbp1-KO mice. In the RRBP1-knockdown Calu-6 cells, a human renin-producing cell line, transmission electron and confocal microscopy revealed that renin was primarily retained in the endoplasmic reticulum and was unable to efficiently target the Golgi apparatus for secretion. CONCLUSIONS RRBP1 deficiency in mice caused hyporeninemic hypoaldosteronism, resulting in lower blood pressure, severe hyperkalemia, and sudden cardiac death. In juxtaglomerular cells, deficiency of RRBP1 reduced renin intracellular trafficking from ER to Golgi apparatus. RRBP1 is a brand-new regulator of blood pressure and potassium homeostasis discovered in this study.
Collapse
Affiliation(s)
- Chu-Hsuan Chiu
- grid.19188.390000 0004 0546 0241Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100 Taiwan ,grid.28665.3f0000 0001 2287 1366Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan
| | - Chin-Feng Hsuan
- grid.414686.90000 0004 1797 2180Division of Cardiology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, 824410 Taiwan ,Division of Cardiology, Department of Internal Medicine, E-Da Dachang Hospital, Kaohsiung, 82445 Taiwan ,grid.411447.30000 0004 0637 1806School of Medicine, College of Medicine, I-Shou University, Kaohsiung, 840203 Taiwan
| | - Shih-Hua Lin
- grid.260565.20000 0004 0634 0356Graduate Institute of Medical Science, National Defense Medical Center, Taipei, 114 Taiwan ,grid.278247.c0000 0004 0604 5314Section of General Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, 111 Taiwan
| | - Yi-Jen Hung
- grid.260565.20000 0004 0634 0356Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, 100 Taiwan
| | - Chii-Min Hwu
- grid.260539.b0000 0001 2059 7017Faculty of Medicine, National Yang-Ming University School of Medicine, Taipei, 112 Taiwan ,grid.278244.f0000 0004 0638 9360Division of Nephrology, Department of Medicine, Tri-Service General Hospital, Taipei, 114 Taiwan
| | - Siow-Wey Hee
- grid.412094.a0000 0004 0572 7815Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100 Taiwan
| | - Shu-Wha Lin
- grid.19188.390000 0004 0546 0241Division of Genomic Medicine, Research Center for Medical Excellence, Transgenic Mouse Models Core, National Taiwan University, Taipei, 100 Taiwan
| | - Sitt-Wai Fong
- grid.28665.3f0000 0001 2287 1366Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan
| | - Patrick Ching-Ho Hsieh
- grid.28665.3f0000 0001 2287 1366Institute of Biomedical Sciences, Academia Sinica, Taipei, 115 Taiwan
| | - Wei-Shun Yang
- grid.19188.390000 0004 0546 0241Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100 Taiwan ,grid.412094.a0000 0004 0572 7815Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Hsin-Chu Branch, Hsin-Chu, 302 Taiwan
| | - Wei-Chou Lin
- grid.412094.a0000 0004 0572 7815Department of Pathology, National Taiwan University Hospital, Taipei, 100 Taiwan
| | - Hsiao-Lin Lee
- grid.412094.a0000 0004 0572 7815Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100 Taiwan
| | - Meng-Lun Hsieh
- grid.412094.a0000 0004 0572 7815Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100 Taiwan ,grid.15276.370000 0004 1936 8091Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL 32610 USA
| | - Wen-Yi Li
- grid.412094.a0000 0004 0572 7815Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, 640 Taiwan
| | - Jou-Wei Lin
- grid.412094.a0000 0004 0572 7815Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, 640 Taiwan
| | - Chih-Neng Hsu
- grid.412094.a0000 0004 0572 7815Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital Yunlin Branch, Yunlin, 640 Taiwan
| | - Vin-Cent Wu
- grid.412094.a0000 0004 0572 7815Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100 Taiwan
| | - Gwo-Tsann Chuang
- grid.19188.390000 0004 0546 0241Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100 Taiwan ,grid.412094.a0000 0004 0572 7815Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, 100 Taiwan
| | - Yi-Cheng Chang
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University, Taipei, 100, Taiwan. .,Institute of Biomedical Sciences, Academia Sinica, Taipei, 115, Taiwan. .,Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan. .,Graduate Institute of Molecular Medicine, National Taiwan University, Taipei, 100, Taiwan.
| | - Lee-Ming Chuang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 100, Taiwan. .,Graduate Institute of Molecular Medicine, National Taiwan University, Taipei, 100, Taiwan. .,Graduate Institute of Clinical Medicine, National Taiwan University, Taipei, 100, Taiwan.
| |
Collapse
|
14
|
Pieri K, Trichia E, Neville MJ, Taylor H, Bennett D, Karpe F, Koivula RW. Polygenic risk in Type III hyperlipidaemia and risk of cardiovascular disease: An epidemiological study in UK Biobank and Oxford Biobank. Int J Cardiol 2023; 373:72-78. [PMID: 36410544 DOI: 10.1016/j.ijcard.2022.11.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Type III hyperlipidaemia (T3HL) is characterised by equimolar increases in plasma triglycerides (TG) and cholesterol in <10% of APOE22 carriers conveying high cardiovascular disease (CVD) risk. We investigate the role of a weighted triglyceride-raising polygenic score (TG.PS) precipitating T3HL. METHODS The TG.PS (restricted to genome-wide significance and weighted by published independent effect estimates) was applied to the Oxford Biobank (OBB, n = 6952) and the UK Biobank (UKB, n = 460,037), to analyse effects on plasma lipid phenotypes. Fasting plasma lipid, lipoprotein biochemistry and NMR lipoprotein profiles were analysed in OBB. CVD prevalence/incidence was examined in UKB. RESULTS One TG.PS standard-deviation (SD) was associated with 13.0% (95% confidence-interval 12.0-14.0%) greater TG in OBB and 15.2% (15.0-15.4%) in UKB. APOE22 carriers had 19.0% (1.0-39.0%) greater TG in UKB. Males were more susceptible to TG.PS effects (4.0% (2.0-6.0%) greater TG with 1 TG.PS SD in OBB, 1.6% (1.3-1.9%) in UKB) than females. There was no interaction between APOE22 and TG.PS, BMI, sex or age on TG. APOE22 carriers had lower apolipoprotein B (apoB) (OBB; -0.35 (-0.29 to -0.40)g/L, UKB; -0.41 (-0.405 to -0.42)g/L). NMR lipoprotein lipid concentrations were discordant to conventional biochemistry in APOE22 carriers. In APOE22 compared with APOE33, CVD was no more prevalent in similarly hypertriglyceridaemic participants (OR 0.97 95%CI 0.76-1.25), but was less prevalent in normolipidaemia (OR 0.81, 95%CI 0.69-0.95); no differences were observed in CVD incidence. CONCLUSIONS TG.PS confers an additive risk for developing T3HL, that is of comparable effect size to conventional risk factors. The protective effect of APOE22 for prevalent CVD is consistent with lower apoB in APOE22 carriers.
Collapse
Affiliation(s)
- Kyriaki Pieri
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, United Kingdom
| | - Eirini Trichia
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Oxford OX3 7LF, United Kingdom; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Oxford OX3 7LF, United Kingdom
| | - Matt J Neville
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, United Kingdom; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford OX4 2PG, United Kingdom
| | - Hannah Taylor
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Oxford OX3 7LF, United Kingdom
| | - Derrick Bennett
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Oxford OX3 7LF, United Kingdom; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford OX4 2PG, United Kingdom; Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Richard Doll Building, Old Road Campus, Oxford OX3 7LF, United Kingdom
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, United Kingdom; NIHR Oxford Biomedical Research Centre, Oxford University Hospitals Foundation Trust, Oxford OX4 2PG, United Kingdom.
| | - Robert W Koivula
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Churchill Hospital, Oxford OX3 7LE, United Kingdom; Genetic and Molecular Epidemiology Unit, Department of Clinical Sciences, Lund University, Skåne University Hospital Malmö, CRC, 91-10, 205 02 Malmö, Sweden.
| |
Collapse
|
15
|
Abstract
Atherosclerotic cardiovascular disease is the leading cause of death globally. Despite its important risk of premature atherosclerosis and cardiovascular disease, familial hypercholesterolemia (FH) is still largely underdiagnosed worldwide. It is one of the most frequently inherited diseases due to mutations, for autosomal dominant forms, in either of the LDLR, APOB, and PCSK9 genes or possibly a few mutations in the APOE gene and, for the rare autosomal forms, in the LDLRAP1 gene. The discovery of the genes implicated in the disease has largely helped to improve the diagnosis and treatment of FH from the LDLR by Brown and Goldstein, as well as the introduction of statins, to PCSK9 discovery in FH by Abifadel et al., and the very rapid availability of PCSK9 inhibitors. In the last two decades, major progress has been made in clinical and genetic diagnostic tools and the therapeutic arsenal against FH. Improving prevention, diagnosis, and treatment and making them more accessible to all patients will help reduce the lifelong burden of the disease.
Collapse
Affiliation(s)
- Marianne Abifadel
- UMR1148, Inserm, Hôpital Bichat-Claude Bernard, 46 rue Henri Huchard, F-75018 Paris, France.,Laboratory of Biochemistry and Molecular Therapeutics (LBTM), Faculty of Pharmacy, Pôle Technologie-Santé, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Catherine Boileau
- UMR1148, Inserm, Hôpital Bichat-Claude Bernard, 46 rue Henri Huchard, F-75018 Paris, France.,Département de Génétique, AP-HP, Hôpital Bichat-Claude Bernard, Paris, France
| |
Collapse
|
16
|
Mitochondrial Genetic Background May Impact Statins Side Effects and Atherosclerosis Development in Familial Hypercholesterolemia. Int J Mol Sci 2022; 24:ijms24010471. [PMID: 36613915 PMCID: PMC9820128 DOI: 10.3390/ijms24010471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022] Open
Abstract
Heredity of familial hypercholesterolemia (FH) can present as a dominant monogenic disorder of polygenic origin or with no known genetic cause. In addition, the variability of the symptoms among individuals or within the same families evidence the potential contribution of additional factors than monogenic mutations that could modulate the development and severity of the disease. In addition, statins, the lipid-lowering drugs which constitute the first-line therapy for the disease, cause associated muscular symptoms in a certain number of individuals. Here, we analyze the evidence of the mitochondrial genetic variation with a special emphasis on the role of CoQ10 to explain this variability found in both disease symptoms and statins side effects. We propose to use mtDNA variants and copy numbers as markers for the cardiovascular disease development of FH patients and to predict potential statin secondary effects and explore new mechanisms to identify new markers of disease or implement personalized medicine strategies for FH therapy.
Collapse
|
17
|
Hovland A, Mundal LJ, Veierød MB, Holven KB, Bogsrud MP, Tell GS, Leren TP, Retterstøl K. The risk of various types of cardiovascular diseases in mutation positive familial hypercholesterolemia; a review. Front Genet 2022; 13:1072108. [PMID: 36561318 PMCID: PMC9763610 DOI: 10.3389/fgene.2022.1072108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 12/12/2022] Open
Abstract
Familial hypercholesterolemia (FH) is a common, inherited disease characterized by high levels of low-density lipoprotein Cholesterol (LDL-C) from birth. Any diseases associated with increased LDL-C levels including atherosclerotic cardiovascular diseases (ASCVDs) would be expected to be overrepresented among FH patients. There are several clinical scoring systems aiming to diagnose FH, however; most individuals who meet the clinical criteria for a FH diagnosis do not have a mutation causing FH. In this review, we aim to summarize the literature on the risk for the various forms of ASCVD in subjects with a proven FH-mutation (FH+). We searched for studies on FH+ and cardiovascular diseases and also included our and other groups published papers on FH + on a wide range of cardiovascular and other diseases of the heart and vessels. FH + patients are at a markedly increased risk of a broad range of ASCVD. Acute myocardial infarction (AMI) is the most common in absolute numbers, but also aortic valve stenosis is by far associated with the highest excess risk. Per thousand patients, we observed 3.6 incident AMI per year compared to 1.9 incident aortic valve stenosis, however, standardized incidence ratio (SIR) for incident AMI was 2.3 compared to 7.9 for incident aortic valve stenosis. Further, occurrence of ischemic stroke seems not to be associated with increased risk in FH+. Clinicians should be aware of the excess risk of almost all kind of ASCVD in FH+, and the neutral risk of stroke need to be studied further in FH + patients.
Collapse
Affiliation(s)
| | - Liv J. Mundal
- The Lipid Clinic, Oslo University Hospital, Oslo, Norway
| | - Marit B. Veierød
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, University of Oslo, Oslo, Norway
| | - Kirsten B. Holven
- Department of Nutrition, University of Oslo, Oslo, Norway,National Advisory Unit on Familial Hypercholesterolemia, Oslo University Hospital, Oslo, Norway
| | - Martin Prøven Bogsrud
- Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital, Oslo, Norway
| | - Grethe S. Tell
- Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway,Division of Mental, Bergen, Norway
| | - Trond P. Leren
- Unit for Cardiac and Cardiovascular Genetics, Oslo University Hospital, Oslo, Norway
| | - Kjetil Retterstøl
- The Lipid Clinic, Oslo University Hospital, Oslo, Norway,Department of Nutrition, University of Oslo, Oslo, Norway,*Correspondence: Kjetil Retterstøl,
| |
Collapse
|
18
|
Arooj S, Rehman SU, Imran A, Almuhaimeed A, Alzahrani AK, Alzahrani A. A Deep Convolutional Neural Network for the Early Detection of Heart Disease. Biomedicines 2022; 10:2796. [PMID: 36359317 PMCID: PMC9687844 DOI: 10.3390/biomedicines10112796] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 08/08/2023] Open
Abstract
Heart disease is one of the key contributors to human death. Each year, several people die due to this disease. According to the WHO, 17.9 million people die each year due to heart disease. With the various technologies and techniques developed for heart-disease detection, the use of image classification can further improve the results. Image classification is a significant matter of concern in modern times. It is one of the most basic jobs in pattern identification and computer vision, and refers to assigning one or more labels to images. Pattern identification from images has become easier by using machine learning, and deep learning has rendered it more precise than traditional image classification methods. This study aims to use a deep-learning approach using image classification for heart-disease detection. A deep convolutional neural network (DCNN) is currently the most popular classification technique for image recognition. The proposed model is evaluated on the public UCI heart-disease dataset comprising 1050 patients and 14 attributes. By gathering a set of directly obtainable features from the heart-disease dataset, we considered this feature vector to be input for a DCNN to discriminate whether an instance belongs to a healthy or cardiac disease class. To assess the performance of the proposed method, different performance metrics, namely, accuracy, precision, recall, and the F1 measure, were employed, and our model achieved validation accuracy of 91.7%. The experimental results indicate the effectiveness of the proposed approach in a real-world environment.
Collapse
Affiliation(s)
- Sadia Arooj
- University Institute of Information Technology, PMAS-Arid Agriculture University, Rawalpindi 46000, Pakistan
| | - Saif ur Rehman
- University Institute of Information Technology, PMAS-Arid Agriculture University, Rawalpindi 46000, Pakistan
| | - Azhar Imran
- Department of Creative Technologies, Faculty of Computing & Artificial Intelligence, Air University, Islamabad 42000, Pakistan
| | - Abdullah Almuhaimeed
- The National Centre for Genomics Technologies and Bioinformatics, King Abdulaziz City for Science and Technology, Riyadh 11442, Saudi Arabia
| | - A. Khuzaim Alzahrani
- Faculty of Applied Medical Sciences, Northern Border University, Arar 91431, Saudi Arabia
| | - Abdulkareem Alzahrani
- Faculty of Computer Science and Information Technology, Al Baha University, Al Baha 65779, Saudi Arabia
| |
Collapse
|
19
|
Kiflen M, Le A, Mao S, Lali R, Narula S, Xie F, Paré G. Cost-Effectiveness of Polygenic Risk Scores to Guide Statin Therapy for Cardiovascular Disease Prevention. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2022; 15:e003423. [PMID: 35904973 DOI: 10.1161/circgen.121.003423] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Atherosclerotic cardiovascular diseases (CVDs) are leading causes of death despite effective therapies and result in unnecessary morbidity and mortality throughout the world. We aimed to investigate the cost-effectiveness of polygenic risk scores (PRS) to guide statin therapy for Canadians with intermediate CVD risk and model its economic outlook. METHODS This cost-utility analysis was conducted using UK Biobank prospective cohort study participants, with recruitment from 2006 to 2010, and at least 10 years of follow-up. We included nonrelated white British-descent participants (n=96 116) at intermediate CVD risk with no prior lipid lowering medication or statin-indicated conditions. A coronary artery disease PRS was used to inform decision to use statins. The effects of statin therapy with and without PRS, as well as CVD events were modelled to determine the incremental cost-effectiveness ratio from a Canadian public health care perspective. We discounted future costs and quality-adjusted life-years by 1.5% annually. RESULTS The optimal economic strategy was when intermediate risk individuals with a PRS in the top 70% are eligible for statins while the lowest 1% are excluded. Base-case analysis at a genotyping cost of $70 produced an incremental cost-effectiveness ratio of $172 906 (143 685 USD) per quality-adjusted life-year. In the probabilistic sensitivity analysis, the intervention has approximately a 50% probability of being cost-effective at $179 100 (148 749 USD) per quality-adjusted life-year. At a $0 genotyping cost, representing individuals with existing genotyping information, PRS-guided strategies dominated standard care when 12% of the lowest PRS individuals were withheld from statins. With improved PRS predictive performance and lower genotyping costs, the incremental cost-effectiveness ratio demonstrates possible cost-effectiveness under thresholds of $150 000 and possibly $50 000 per quality-adjusted life-year. CONCLUSIONS This study suggests that using PRS alongside existing guidelines might be cost-effective for CVD. Stronger predictiveness combined with decreased cost of PRS could further improve cost-effectiveness, providing an economic basis for its inclusion into clinical care.
Collapse
Affiliation(s)
- Michel Kiflen
- Department of Medicine, University of Toronto, Toronto (M.K.).,Population Health Research Institute (M.K., A.L., S.M., R.L., S.N., G.P.), McMaster University, Hamilton, Ontario, Canada
| | - Ann Le
- Population Health Research Institute (M.K., A.L., S.M., R.L., S.N., G.P.), McMaster University, Hamilton, Ontario, Canada.,Department of Medical Sciences (A.L.), McMaster University, Hamilton, Ontario, Canada
| | - Shihong Mao
- Population Health Research Institute (M.K., A.L., S.M., R.L., S.N., G.P.), McMaster University, Hamilton, Ontario, Canada
| | - Ricky Lali
- Population Health Research Institute (M.K., A.L., S.M., R.L., S.N., G.P.), McMaster University, Hamilton, Ontario, Canada.,Department of Health Research Methods, Evidence, and Impact (R.L., S.N., F.X., G.P.), McMaster University, Hamilton, Ontario, Canada
| | - Sukrit Narula
- Population Health Research Institute (M.K., A.L., S.M., R.L., S.N., G.P.), McMaster University, Hamilton, Ontario, Canada.,Department of Internal Medicine, Yale University, New Haven, CT (S.N.)
| | - Feng Xie
- Department of Health Research Methods, Evidence, and Impact (R.L., S.N., F.X., G.P.), McMaster University, Hamilton, Ontario, Canada
| | - Guillaume Paré
- Population Health Research Institute (M.K., A.L., S.M., R.L., S.N., G.P.), McMaster University, Hamilton, Ontario, Canada.,Department of Health Research Methods, Evidence, and Impact (R.L., S.N., F.X., G.P.), McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine (G.P.), McMaster University, Hamilton, Ontario, Canada.,Thrombosis & Atherosclerosis Research Institute (G.P.), McMaster University, Hamilton, Ontario, Canada.,McMaster University, Hamilton, Ontario, Canada (G.P.)
| |
Collapse
|
20
|
Park S, Jang M, Park MY, Kim J, Shin S. Interactive effects of the low-carbohydrate diet score and genetic risk score on Hypo-HDL-cholesterolemia among Korean adults: A cross-sectional analysis from the Ansan and Ansung Study of the Korean Genome and Epidemiology Study. Food Sci Nutr 2022; 10:3106-3116. [PMID: 36171780 PMCID: PMC9469851 DOI: 10.1002/fsn3.2909] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
This cross-sectional study investigated the interaction between the genetic risk score (GRS) and abnormal high-density lipoprotein (HDL) cholesterol lipid levels, which are modified by low-carbohydrate diets (LCDs) and their effects on the prevalence of hypo-HDL-cholesterolemia (hypo-HDL-C) in Korean adults. Baseline data were obtained from the Ansan and Ansung study of the Korean Genome and Epidemiology Study (KoGES), conducted from 2001 to 2002, that targeted 8,314 Korean adults aged 40-69 years, including old men (47.6%) and women (52.4%), and whole genomic single nucleotide polymorphism (SNP) genotyping was performed. We identified 18 SNPs significantly associated with hypo-HDL-C in the proximity of several genes, including LPL, APOA5, LIPC, and CETP, and calculated the GRS. The low-carbohydrate diet score (LCDS) was calculated on the basis of energy intake information from food frequency questionnaires. Furthermore, we performed multivariable-adjusted logistic modeling to examine the odds ratio (OR) for hypo-HDL-C across tertiles of LCDS and GRS, adjusted for several covariates. Among participants in the highest GRS tertile, those in the highest tertile of the LCDS had a significantly lower risk of hypo-HDL-C (OR: 0.759, 95% CI (confidence interval): 0.625-0.923) than those in the lowest tertile of the LCDS. In the joint effect model, the group with the lowest GRS and highest LCDS was found to have the lowest risk of hypo-HDL-C prevalence. This study suggests that individuals with a high genetic risk for low HDL concentrations may have a beneficial effect on a lower intake of carbohydrates.
Collapse
Affiliation(s)
- SoHyun Park
- Department of Food and NutritionChung‐Ang UniversityGyeonggi‐doKorea
| | - Min‐Jae Jang
- Department of Animal Science and TechnologyChung‐Ang UniversityGyeonggi‐doKorea
| | - Min Young Park
- Department of Molecular PathobiologyNYU College of DentistryNew YorkNew YorkUSA
| | - Jun‐Mo Kim
- Department of Animal Science and TechnologyChung‐Ang UniversityGyeonggi‐doKorea
| | - Sangah Shin
- Department of Food and NutritionChung‐Ang UniversityGyeonggi‐doKorea
| |
Collapse
|
21
|
Hill EJ, Robak LA, Al-Ouran R, Deger J, Fong JC, Vandeventer PJ, Schulman E, Rao S, Saade H, Savitt JM, von Coelln R, Desai N, Doddapaneni H, Salvi S, Dugan-Perez S, Muzny DM, McGuire AL, Liu Z, Gibbs RA, Shaw C, Jankovic J, Shulman LM, Shulman JM. Genome Sequencing in the Parkinson Disease Clinic. Neurol Genet 2022; 8:e200002. [PMID: 35747619 PMCID: PMC9210549 DOI: 10.1212/nxg.0000000000200002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 04/22/2022] [Indexed: 11/18/2022]
Abstract
Background and Objectives Genetic variants affect both Parkinson disease (PD) risk and manifestations. Although genetic information is of potential interest to patients and clinicians, genetic testing is rarely performed during routine PD clinical care. The goal of this study was to examine interest in comprehensive genetic testing among patients with PD and document reactions to possible findings from genome sequencing in 2 academic movement disorder clinics. Methods In 203 subjects with PD (age = 63 years, 67% male), genome sequencing was performed and filtered using a custom panel, including 49 genes associated with PD, parkinsonism, or related disorders, as well as a 90-variant PD genetic risk score. Based on the results, 231 patients (age = 67 years, 63% male) were surveyed on interest in genetic testing and responses to vignettes covering (1) familial risk of PD (LRRK2); (2) risk of PD dementia (GBA); (3) PD genetic risk score; and (4) secondary, medically actionable variants (BRCA1). Results Genome sequencing revealed a LRRK2 variant in 3% and a GBA risk variant in 10% of our clinical sample. The genetic risk score was normally distributed, identifying 41 subjects with a high risk of PD. Medically actionable findings were discovered in 2 subjects (1%). In our survey, the majority (82%) responded that they would share a LRRK2 variant with relatives. Most registered unchanged or increased interest in testing when confronted with a potential risk for dementia or medically actionable findings, and most (75%) expressed interest in learning their PD genetic risk score. Discussion Our results highlight broad interest in comprehensive genetic testing among patients with PD and may facilitate integration of genome sequencing in clinical practice.
Collapse
Affiliation(s)
| | | | - Rami Al-Ouran
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Jennifer Deger
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Jamie C. Fong
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Paul Jerrod Vandeventer
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Emily Schulman
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Sindhu Rao
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Hiba Saade
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Joseph M. Savitt
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Rainer von Coelln
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Neeja Desai
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Harshavardhan Doddapaneni
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Sejal Salvi
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Shannon Dugan-Perez
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Donna M. Muzny
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Amy L. McGuire
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Zhandong Liu
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Richard A. Gibbs
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Chad Shaw
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Joseph Jankovic
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Lisa M. Shulman
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| | - Joshua M. Shulman
- From the Department of Neurology (E.J.H., S.R., H.S., J.J., J.M. Shulman), and Parkinson's Disease Center and Movement Disorders Clinic (E.J.H., C.S., J.J., J.M. Shulman), Baylor College of Medicine, Houston, TX. E.J. Hill is now with Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH; Department of Molecular and Human Genetics (L.A.R., J.C.F., P.J.V., R.A.G., J.M. Shulman), Department of Pediatrics (R.A.-O., Z.L.), and Department of Neuroscience (J.D., J.M. Shulman), Baylor College of Medicine, Houston, TX; Department of Neurology (E.S., J.M. Savitt, R.v.C., N.D., L.M.S.), University of Maryland School of Medicine, Baltimore; Center for Alzheimer's and Neurodegenerative Diseases (H.S., A.L.M., Z.L., J.M. Shulman), Human Genome Sequencing Center (H.D., S.S., S.D.-P., D.M.M., R.A.G.), and Center for Medical Ethics and Health Policy (A.L.M.), Baylor College of Medicine, Houston, TX; and Jan and Dan Duncan Neurological Research Institute (Z.L., J.M. Shulman), Texas Children's Hospital, Houston
| |
Collapse
|
22
|
Lima IR, Tada MT, Oliveira TG, Jannes CE, Bensenor I, Lotufo PA, Santos RD, Krieger JE, Pereira AC. Polygenic risk score for hypercholesterolemia in a Brazilian familial hypercholesterolemia cohort. ATHEROSCLEROSIS PLUS 2022; 49:47-55. [PMID: 36644206 PMCID: PMC9833269 DOI: 10.1016/j.athplu.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/03/2022] [Accepted: 06/24/2022] [Indexed: 01/18/2023]
Abstract
Background and aims Familial hypercholesterolemia (FH) is a genetic disorder characterized by high levels of LDL-C leading to premature cardiovascular disease (CAD). Only about 40% of individuals with a clinical diagnosis of FH have a causative genetic variant identified, and a proportion of genetically negative cases may have a polygenic cause rather than a still unidentified monogenic cause. This work aims to evaluate and validate the role of a polygenic risk score (PRS) associated with hypercholesterolemia in a Brazilian FH cohort and its clinical implications. Methods We analyzed a previously derived PRS of 12 and 6 SNPs (Single Nucleotide Polymorphism) in 684 FH individuals (491 mutation-negative [FH/M-], 193 mutation-positive [FH/M+]) and in 1605 controls. Coronary artery calcium (CAC) score was also evaluated. Results The PRS was independently associated with LDL-C in control individuals (p < 0.001). Within this group, in individuals in the highest quartile of the 12 SNPs PRS, the odds ratio for CAC score >100 was 1.7 (95% CI: 1.01-2.88, p = 0.04) after adjustment for age and sex. Subjects in the FH/M- group had the highest mean score in both 12 and 6 SNPs PRS (38.25 and 27.82, respectively) when compared to the other two groups (p = 2.2 × 10-16). Both scores were also higher in the FH/M+ group (36.48 and 26.26, respectively) when compared to the control group (p < 0.001 for the two scores) but inferior to the FH/M- group. Within FH individuals, the presence of a higher PRS score was not associated with LDL-C levels or with CAD risk. Conclusion A higher PRS is associated with significantly higher levels of LDL-C and it is independently associated with higher CAC in the Brazilian general population. A polygenic cause can explain a fraction of FH/M- individuals but does not appear to be a modulator of the clinical phenotype among FH individuals, regardless of mutation status.
Collapse
Affiliation(s)
- Isabella Ramos Lima
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil,Corresponding author.
| | - Mauricio Teruo Tada
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Theo G.M. Oliveira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Cinthia Elim Jannes
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Isabela Bensenor
- Center for Clinical and Epidemiologic Research, University of São Paulo, São Paulo, Brazil
| | - Paulo A. Lotufo
- Center for Clinical and Epidemiologic Research, University of São Paulo, São Paulo, Brazil
| | - Raul D. Santos
- Lipid Clinic, Heart Institute (InCor), University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Jose E. Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Alexandre C. Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil,Genetics Department, Harvard Medical School, Boston, MA, USA,Corresponding author. Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
23
|
Polygenic risk scores for cardiovascular disease prediction in the clinical practice: Are we there? Atherosclerosis 2021; 340:46-47. [PMID: 34895918 DOI: 10.1016/j.atherosclerosis.2021.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022]
|
24
|
Paquette M, Bernard S, Cariou B, Hegele RA, Genest J, Trinder M, Brunham LR, Béliard S, Baass A. Familial Hypercholesterolemia-Risk-Score: A New Score Predicting Cardiovascular Events and Cardiovascular Mortality in Familial Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2021; 41:2632-2640. [PMID: 34433300 DOI: 10.1161/atvbaha.121.316106] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Objective: Familial hypercholesterolemia (FH) is associated with a high risk of premature atherosclerotic cardiovascular disease (ASCVD). However, this risk is highly heterogeneous and current risk prediction algorithms for FH suffer from limitations. The primary objective of this study was to develop a score predicting incident ASCVD events over 10 years in a large multinational FH cohort. The secondary objective was to investigate the prediction of major adverse cardiovascular events and cardiovascular mortality using this score.
Approach and Results: We prospectively followed 3881 patients with adult heterozygous FH with no prior history of ASCVD (32 361 person-years of follow-up) from 5 registries in Europe and North America. The FH-Risk-Score incorporates 7 clinical variables: sex, age, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, hypertension, smoking, and lipoprotein (a) (Lp(a)) with a Harrell C-index for 10-year ASCVD event of 0.75, which was superior to the SAFEHEART-RE (Spanish Familial Hypercholesterolemia Cohort; 0.69). Subjects with an elevated FH-Risk-Score had decreases in 10-year ASCVD-free survival, 10-year major adverse cardiovascular event-free survival, and 30-year survival for CV mortality compared with the low-risk group, with hazard ratios of 5.52 (3.94-7.73), 4.64 (2.66-8.11), and 10.73 (2.51-45.79), respectively. The FH-Risk-Score showed a similar performance in subjects with and without an FH-causing mutation.
Conclusions: The FH-Risk-Score is a stronger predictor of future ASCVD than the SAFEHEART-RE and was developed in FH subjects with no prior cardiovascular event. Furthermore, the FH-Risk-Score is the first score to predict CV death and could offer personalized cardiovascular risk assessment and treatment for patients with FH. Future studies are required to validate the FH-Risk-Score in different ethnic groups.
Collapse
Affiliation(s)
- Martine Paquette
- Lipids, Nutrition, and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Québec, Canada (M.P., S. Bernard, A.B.)
| | - Sophie Bernard
- Lipids, Nutrition, and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Québec, Canada (M.P., S. Bernard, A.B.)
- Department of Medicine, Division of Endocrinology, Université de Montreal, Québec, Canada (S. Bernard)
| | - Bertrand Cariou
- L'institut du thorax, Department of Endocrinology, UNIV Nantes, CNRS, Inserm, CHU Nantes, France (B.C.)
| | - Robert A Hegele
- Departments of Medicine and Biochemistry, and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, Ontario, Canada (R.A.H.)
| | - Jacques Genest
- Research Institute of the McGill University Health Centre, Québec, Canada (J.G.)
| | - Mark Trinder
- Faculty of Medicine, University of British Columbia, Centre for Heart and Lung Innovation, Department of Medicine, University of British Columbia, Canada (M.T., L.R.B.)
| | - Liam R Brunham
- Faculty of Medicine, University of British Columbia, Centre for Heart and Lung Innovation, Department of Medicine, University of British Columbia, Canada (M.T., L.R.B.)
| | - Sophie Béliard
- Aix Marseille University, INSERM, INRA, C2VN, Department of Nutrition, Metabolic Diseases, Endocrinology, La Conception Hospital, Marseille, France (S. Béliard)
| | - Alexis Baass
- Lipids, Nutrition, and Cardiovascular Prevention Clinic of the Montreal Clinical Research Institute, Québec, Canada (M.P., S. Bernard, A.B.)
- Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Québec, Canada (A.B.)
| |
Collapse
|
25
|
Paquette M, Fantino M, Bernard S, Baass A. Paternal inheritance predicts earlier cardiovascular event onset in patients with familial hypercholesterolemia. Atherosclerosis 2021; 329:9-13. [PMID: 34157652 DOI: 10.1016/j.atherosclerosis.2021.06.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 05/20/2021] [Accepted: 06/08/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND AND AIMS Familial hypercholesterolemia (FH) is a genetic disease, with an autosomal codominant inheritance, predisposing to premature atherosclerotic cardiovascular disease (ASCVD). Paternal or maternal inheritance of the FH-causing mutation may affect the FH phenotype in offspring, but the effect of the genetic transmission on cardiovascular disease risk remains to be established. The aim of the present study is to compare the incidence of cardiovascular events between patients with maternal vs paternal inheritance of familial hypercholesterolemia. METHODS We prospectively studied 725 genetically-confirmed FH patients (33,805 person-years), including 268 with maternal inheritance and 321 with paternal inheritance of the mutation. ASCVD was defined as angina, myocardial infarction, coronary angioplasty, coronary bypass surgery, claudication, peripheral angioplasty, peripheral arterial surgery, transient ischemic attack, stroke, carotid endarterectomy and CV death. Cox-proportional hazard models and Kaplan-Meier analysis were used to compare the two groups. RESULTS Before 50 years of age, paternal inheritance of FH was associated with a 1.5-fold increased risk for ASCVD, as compared to maternal inheritance (HR 1.59, 95% CI 1.11-2.28, p = 0.01). This association remained significant after adjusting for confounding factors (HR 1.49, 95% CI 1.00-2.23, p = 0.05). The age of first ASCVD event was also significantly lower in the paternal inheritance group (42 years) than in the maternal inheritance group (46 years), p = 0.02. CONCLUSIONS This study suggests that paternal inheritance of the FH-causing mutation was associated with an earlier cardiovascular event onset compared to maternal inheritance. The mechanisms behind these findings remain to be established.
Collapse
Affiliation(s)
- Martine Paquette
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Québec, Canada.
| | - Manon Fantino
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Québec, Canada
| | - Sophie Bernard
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Québec, Canada; Department of Medicine, Division of Endocrinology, Université de Montreal, Québec, Canada
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Québec, Canada; Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Québec, Canada.
| |
Collapse
|
26
|
Goodrich JK, Singer-Berk M, Son R, Sveden A, Wood J, England E, Cole JB, Weisburd B, Watts N, Caulkins L, Dornbos P, Koesterer R, Zappala Z, Zhang H, Maloney KA, Dahl A, Aguilar-Salinas CA, Atzmon G, Barajas-Olmos F, Barzilai N, Blangero J, Boerwinkle E, Bonnycastle LL, Bottinger E, Bowden DW, Centeno-Cruz F, Chambers JC, Chami N, Chan E, Chan J, Cheng CY, Cho YS, Contreras-Cubas C, Córdova E, Correa A, DeFronzo RA, Duggirala R, Dupuis J, Garay-Sevilla ME, García-Ortiz H, Gieger C, Glaser B, González-Villalpando C, Gonzalez ME, Grarup N, Groop L, Gross M, Haiman C, Han S, Hanis CL, Hansen T, Heard-Costa NL, Henderson BE, Hernandez JMM, Hwang MY, Islas-Andrade S, Jørgensen ME, Kang HM, Kim BJ, Kim YJ, Koistinen HA, Kooner JS, Kuusisto J, Kwak SH, Laakso M, Lange L, Lee JY, Lee J, Lehman DM, Linneberg A, Liu J, Loos RJF, Lyssenko V, Ma RCW, Martínez-Hernández A, Meigs JB, Meitinger T, Mendoza-Caamal E, Mohlke KL, Morris AD, Morrison AC, Ng MCY, Nilsson PM, O'Donnell CJ, Orozco L, Palmer CNA, Park KS, Post WS, Pedersen O, Preuss M, Psaty BM, Reiner AP, Revilla-Monsalve C, Rich SS, Rotter JI, Saleheen D, Schurmann C, Sim X, Sladek R, Small KS, So WY, Spector TD, Strauch K, Strom TM, Tai ES, Tam CHT, Teo YY, Thameem F, Tomlinson B, Tracy RP, Tuomi T, Tuomilehto J, Tusié-Luna T, van Dam RM, Vasan RS, Wilson JG, Witte DR, Wong TY, Burtt NP, Zaitlen N, McCarthy MI, Boehnke M, Pollin TI, Flannick J, Mercader JM, O'Donnell-Luria A, Baxter S, Florez JC, MacArthur DG, Udler MS. Determinants of penetrance and variable expressivity in monogenic metabolic conditions across 77,184 exomes. Nat Commun 2021; 12:3505. [PMID: 34108472 PMCID: PMC8190084 DOI: 10.1038/s41467-021-23556-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 04/27/2021] [Indexed: 11/16/2022] Open
Abstract
Hundreds of thousands of genetic variants have been reported to cause severe monogenic diseases, but the probability that a variant carrier develops the disease (termed penetrance) is unknown for virtually all of them. Additionally, the clinical utility of common polygenetic variation remains uncertain. Using exome sequencing from 77,184 adult individuals (38,618 multi-ancestral individuals from a type 2 diabetes case-control study and 38,566 participants from the UK Biobank, for whom genotype array data were also available), we apply clinical standard-of-care gene variant curation for eight monogenic metabolic conditions. Rare variants causing monogenic diabetes and dyslipidemias display effect sizes significantly larger than the top 1% of the corresponding polygenic scores. Nevertheless, penetrance estimates for monogenic variant carriers average 60% or lower for most conditions. We assess epidemiologic and genetic factors contributing to risk prediction in monogenic variant carriers, demonstrating that inclusion of polygenic variation significantly improves biomarker estimation for two monogenic dyslipidemias.
Collapse
Affiliation(s)
- Julia K Goodrich
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Moriel Singer-Berk
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rachel Son
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Abigail Sveden
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jordan Wood
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eleina England
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joanne B Cole
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ben Weisburd
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Nick Watts
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lizz Caulkins
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Peter Dornbos
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ryan Koesterer
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Zachary Zappala
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Haichen Zhang
- School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Kristin A Maloney
- School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Andy Dahl
- Department of Neurology, UCLA, Los Angeles, CA, USA
| | | | - Gil Atzmon
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
- Faculty of Natural Science, University of Haifa, Haifa, Israel
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | | | - Nir Barzilai
- Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
- Department of Genetics, Albert Einstein College of Medicine, New York, NY, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville and Edinburg, TX, USA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Lori L Bonnycastle
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Erwin Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Donald W Bowden
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | - John C Chambers
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - Edmund Chan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
| | - Juliana Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Yoon Shin Cho
- Department of Biomedical Science, Hallym University, Chuncheon, South Korea
| | | | - Emilio Córdova
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Adolfo Correa
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ralph A DeFronzo
- Department of Medicine, University of Texas Health San Antonio (aka University of Texas Health Science Center at San Antonio), San Antonio, TX, USA
| | - Ravindranath Duggirala
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville and Edinburg, TX, USA
| | - Josée Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Ma Eugenia Garay-Sevilla
- Department of Medical Science, División of Health Science, University of Guanjuato. Campus León. León, Guanjuato, Mexico
| | | | - Christian Gieger
- Research Unit Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Benjamin Glaser
- Endocrinology and Metabolism Service, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Clicerio González-Villalpando
- Unidad de Investigacion en Diabetes y Riesgo Cardiovascular, Instituto Nacional de Salud Publica, Cuernavaca, Mexico
| | | | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Leif Groop
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Genetics Finland, University of Helsinki, Helsinki, Finland
| | - Myron Gross
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Christopher Haiman
- Department of Preventive Medicine, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Sohee Han
- Division of Genome Research, Center for Genome Science, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Craig L Hanis
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nancy L Heard-Costa
- Boston University and National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Brian E Henderson
- Department of Preventive Medicine, Keck School of Medicine of USC, Los Angeles, CA, USA
| | - Juan Manuel Malacara Hernandez
- Department of Medical Science, División of Health Science, University of Guanjuato. Campus León. León, Guanjuato, Mexico
| | - Mi Yeong Hwang
- Division of Genome Research, Center for Genome Science, National Institute of Health, Chungcheongbuk-do, South Korea
| | | | - Marit E Jørgensen
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
- Greenland Centre for Health Research, University of Greenland, Nuuk, Greenland
| | - Hyun Min Kang
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Bong-Jo Kim
- Division of Genome Research, Center for Genome Science, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Young Jin Kim
- Division of Genome Research, Center for Genome Science, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Heikki A Koistinen
- Department of Public Health Solutions, Finnish Institute for Health and Welfare, Helsinki, Finland
- University of Helsinki and Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Jaspal Singh Kooner
- Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, London, UK
- MRC-PHE Centre for Environment and Health, Imperial College London, London, UK
- Imperial College Healthcare NHS Trust, Imperial College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Soo-Heon Kwak
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Leslie Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Jong-Young Lee
- Oneomics Soonchunhyang Mirae Medical Center, Bucheon-si Gyeonggi-do, Republic of Korea
| | - Juyoung Lee
- Division of Genome Research, Center for Genome Science, National Institute of Health, Chungcheongbuk-do, South Korea
| | - Donna M Lehman
- Department of Medicine, University of Texas Health San Antonio (aka University of Texas Health Science Center at San Antonio), San Antonio, TX, USA
| | - Allan Linneberg
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Clinical Experimental Research, Rigshospitalet, Copenhagen, Denmark
| | - Jianjun Liu
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Genome Institute of Singapore, Agency for Science Technology and Research, Singapore, Singapore
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Ichan School of Medicine at Mount Sinai, New York, NY, USA
| | - Valeriya Lyssenko
- Centro de Estudios en Diabetes, Mexico City, Mexico
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | | | - James B Meigs
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Thomas Meitinger
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | | | - Karen L Mohlke
- Department of Genetics, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| | - Andrew D Morris
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Biostatistics, University of Liverpool, Liverpool, UK
| | - Alanna C Morrison
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Maggie C Y Ng
- Center for Diabetes Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Center for Genomics and Personalized Medicine Research, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Peter M Nilsson
- Department of Clinical Sciences, Medicine, Lund University, Malmö, Sweden
| | - Christopher J O'Donnell
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Section of Cardiology, Department of Medicine, VA Boston Healthcare, Boston, MA, USA
- Brigham and Women's Hospital, Boston, MA, USA
- Intramural Administration Management Branch, National Heart Lung and Blood Institute, NIH, Framingham, MA, USA
| | - Lorena Orozco
- Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Colin N A Palmer
- Pat Macpherson Centre for Pharmacogenetics and Pharmacogenomics, University of Dundee, Dundee, UK
| | - Kyong Soo Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, South Korea
| | - Wendy S Post
- Division of Cardiology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Michael Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology, and Health Services, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Research Institute, Seattle, WA, USA
| | | | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation (formerly Los Angeles Biomedical Research Institute) at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Danish Saleheen
- Division of Translational Medicine and Human Genetics, University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA, USA
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Digital Health Center, Hasso Plattner Institute, University of Potsdam, Prof.-Dr.-Helmert-Str. 2-3, Potsdam, Germany
- Hasso Plattner Institute for Digital Health at Mount Sinai, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Rob Sladek
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McGill University, Montreal, QC, Canada
- McGill University and Génome Québec Innovation Centre, Montreal, QC, Canada
| | - Kerrin S Small
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Wing Yee So
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
| | - Timothy D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum Munchen, German Research Center for Environmental Health, Neuherberg, Germany
- Institute for Medical Informatics Biometry and Epidemiology, Ludwig-Maximilians University, Munich, Germany
| | - Tim M Strom
- Institute of Human Genetics, Technical University of Munich, Munich, Germany
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - E Shyong Tai
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
| | - Claudia H T Tam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, China
| | - Yik Ying Teo
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Statistics and Applied Probability, National University of Singapore, Singapore, Singapore
| | - Farook Thameem
- Department of Biochemistry, Faculty of Medicine, Health Science Center, Kuwait University, Safat, Kuwait
| | - Brian Tomlinson
- Faculty of Medicine, Macau University of Science & Technology, Macau, China
| | - Russell P Tracy
- Department of Pathology and Laboratory Medicine, The Robert Larner M.D. College of Medicine, University of Vermont, Burlington, VT, USA
- Department of Biochemistry, The Robert Larner M.D. College of Medicine, University of Vermont, Burlington, VT, USA
| | - Tiinamaija Tuomi
- Department of Clinical Sciences, Diabetes and Endocrinology, Lund University Diabetes Centre, Malmö, Sweden
- Institute for Molecular Genetics Finland, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Centre, Helsinki, Finland
- Department of Endocrinology, Abdominal Centre, Helsinki University Hospital, Helsinki, Finland
- Research Programs Unit, Clinical and Molecular Medicine, University of Helsinki, Helsinki, Finland
| | - Jaakko Tuomilehto
- Public Health Promotion Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Saudi Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of International Health, National School of Public Health, Instituto de Salud Carlos III, Madrid, Spain
| | - Teresa Tusié-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
- Departamento de Medicina Genómica y Toxiología Ambiental, Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico
| | - Rob M van Dam
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore, Singapore
- Department of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - Ramachandran S Vasan
- Boston University and National Heart Lung and Blood Institute's Framingham Heart Study, Framingham, MA, USA
- Preventive Medicine & Epidemiology, and Cardiovascular Medicine, Medicine, Boston University School of Medicine, and Epidemiology, Boston University School of Public health, Boston, MA, USA
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daniel R Witte
- Department of Public Health, Aarhus University, Aarhus, Denmark
- Danish Diabetes Academy, Odense, Denmark
| | - Tien-Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Singapore
- Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Noël P Burtt
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Noah Zaitlen
- Department of Neurology, UCLA, Los Angeles, CA, USA
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Genentech, South San Francisco, CA, USA
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Toni I Pollin
- School of Medicine, University of Maryland Baltimore, Baltimore, MD, USA
| | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Josep M Mercader
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Anne O'Donnell-Luria
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Samantha Baxter
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jose C Florez
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Daniel G MacArthur
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Centre for Population Genomics, Garvan Institute of Medical Research, UNSW Sydney, Sydney, NSW, Australia
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Miriam S Udler
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Fantino M, Paquette M, Bernard S, Baass A. ANKS1A genotype predicts cardiovascular events in patients with familial hypercholesterolemia. J Clin Lipidol 2021; 15:602-607. [PMID: 34130940 DOI: 10.1016/j.jacl.2021.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND The rs17609940 variant of the ANKS1A gene has been associated with coronary artery disease (CAD) risk in genome-wide association studies (GWAS), but no study has yet replicated this association in familial hypercholesterolemia (FH) population. OBJECTIVE The aim of this study is to validate the association between the rs17609940 genotype and incident major adverse cardiovascular events (MACE) in a cohort of genetically-confirmed FH patients. METHODS This association study includes 725 genetically-confirmed FH patients with a median observation period of 50 years (33 805 person-years). MACE were defined as either myocardial infarction (MI), stroke, coronary revascularization, hospital admission for unstable angina and cardiovascular disease (CVD) death. The rs17609940 genotype was imputed with an imputation quality of 0.831 following an exome chip genotyping method (Illumina). RESULTS The cohort comprised 469 subjects with GG genotype, 218 subjects with CG genotype and 38 subjects with CC genotype. All baseline characteristics were balanced between the three groups. The CC genotype of rs17609940 was associated with a significant lower risk of incident MACE compared to GG and GC carriers in a recessive model (HR 0.30, 95% CI 0.11-0.82, p=0.02). Even after correction for confounding cardiovascular risk factors, the association between the ANKS1A polymorphism and incident MACE remained strongly significant. CONCLUSIONS We demonstrated that the rs17609940 SNP of the ANKS1A gene is associated with the risk of incident MACE in FH subjects. The exact mechanism underlying this association remains to be clarified.
Collapse
Affiliation(s)
- Manon Fantino
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Québec, Canada
| | - Martine Paquette
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Québec, Canada
| | - Sophie Bernard
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Québec, Canada; Department of Medicine, Division of Endocrinology, Université de Montreal, Québec, Canada
| | - Alexis Baass
- Genetic Dyslipidemias Clinic of the Montreal Clinical Research Institute, Québec, Canada; Department of Medicine, Divisions of Experimental Medicine and Medical Biochemistry, McGill University, Québec, Canada.
| |
Collapse
|
28
|
Polygenic risk scores for low-density lipoprotein cholesterol and familial hypercholesterolemia. J Hum Genet 2021; 66:1079-1087. [PMID: 33967275 DOI: 10.1038/s10038-021-00929-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 12/19/2022]
Abstract
Familial hypercholesterolemia (FH) is an autosomal dominant monogenic disorder characterized by elevated levels of low-density lipoprotein cholesterol (LDL-C) and an increased risk of premature coronary artery disease (CAD). Recently, it has been shown that a high polygenic risk score (PRS) could be an independent risk factor for CAD in FH patients of European ancestry. However, it is uncertain whether PRS is also useful for risk stratification of FH patients in East Asia. We recruited and genotyped clinically diagnosed FH (CDFH) patients from the Kanazawa University Mendelian Disease FH registry and controls from the Shikamachi Health Improvement Practice genome cohort in Japan. We calculated PRS from 3.6 million variants of each participant (imputed from the 1000 Genome phase 3 Asian dataset) for LDL-C (PRSLDLC) using a genome-wide association study summary statistic from the BioBank Japan Project. We assessed the association of PRSLDLC with LDL-C and CAD among and within monogenic FH, mutation negative CDFH, and controls. We tested a total of 1223 participants (376 FH patients, including 173 with monogenic FH and 203 with mutation negative CDFH, and 847 controls) for the analyses. PRSLDLC was significantly higher in mutation negative CDFH patients than in controls (p = 3.1 × 10-13). PRSLDLC was also significantly linked to LDL-C in controls (p trend = 3.6 × 10-4) but not in FH patients. Moreover, we could not detect any association between PRSLDLC and CAD in any of the groups. In conclusion, mutation negative CDFH patients demonstrated significantly higher PRSLDLC than controls. However, PRSLDLC may have little additional effect on LDL-C and CAD among FH patients.
Collapse
|
29
|
Precision Medicine Approaches to Vascular Disease: JACC Focus Seminar 2/5. J Am Coll Cardiol 2021; 77:2531-2550. [PMID: 34016266 DOI: 10.1016/j.jacc.2021.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/31/2021] [Accepted: 04/02/2021] [Indexed: 12/16/2022]
Abstract
In this second of a 5-part Focus Seminar series, we focus on precision medicine in the context of vascular disease. The most common vascular disease worldwide is atherosclerosis, which is the primary cause of coronary artery disease, peripheral vascular disease, and a large proportion of strokes and other disorders. Atherosclerosis is a complex genetic disease that likely involves many hundreds to thousands of single nucleotide polymorphisms, each with a relatively modest effect for causing disease. Conversely, although less prevalent, there are many vascular disorders that typically involve only a single genetic change, but these changes can often have a profound effect that is sufficient to cause disease. These are termed "Mendelian vascular diseases," which include Marfan and Loeys-Dietz syndromes. Given the very different genetic basis of atherosclerosis versus Mendelian vascular diseases, this article was divided into 2 parts to cover the most promising precision medicine approaches for these disease types.
Collapse
|
30
|
Matthews LJ, Lebowitz MS, Ottman R, Appelbaum PS. Pygmalion in the Genes? On the potentially negative impacts of polygenic scores for educational attainment. SOCIAL PSYCHOLOGY OF EDUCATION 2021; 24:789-808. [PMID: 39850644 PMCID: PMC11756904 DOI: 10.1007/s11218-021-09632-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 04/01/2021] [Indexed: 02/08/2023]
Abstract
Polygenic scores for educational attainment and related variables, such as IQ and "mathematical ability" are now readily available via direct-to-consumer genetic testing companies. Some researchers are even proposing the use of genetic tests in educational settings via "precision education," in which individualized student education plans would be tailored to polygenic scores. The potential psychosocial impacts of polygenic scores for traits and outcomes relevant to education, however, have not been assessed. In online experiments, we asked participants to imagine hypothetical situations in which they or their classmates had recently received polygenic scores for educational attainment. Participants prompted to answer multi-choice questions as though they had received their own low-percentile score, compared to a control condition, scored significantly lower on measures of self-esteem and of self-perceived competence, academic efficacy, and educational potential. Similarly, those asked to evaluate a hypothetical classmate as though the classmate had received a low-percentile score attributed significantly lower academic efficacy and educational potential, compared to a control condition. Through possible mechanisms of stigma and self-fulfilling prophecies, our results highlight the potential psychosocial harms of exposure to low-percentile polygenic scores for educational attainment.
Collapse
Affiliation(s)
- Lucas J. Matthews
- Department of Medical Humanities & Ethics, Columbia University Irving Medical Center
| | | | - Ruth Ottman
- Department of Psychiatry, Columbia University
- Departments of Epidemiology and Neurology, and the G.H. Sergievsky Center, Columbia University Irving Medical Center
| | | |
Collapse
|
31
|
Trinder M, Brunham LR. Polygenic scores for dyslipidemia: the emerging genomic model of plasma lipoprotein trait inheritance. Curr Opin Lipidol 2021; 32:103-111. [PMID: 33395106 DOI: 10.1097/mol.0000000000000737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Contemporary polygenic scores, which summarize the cumulative contribution of millions of common single-nucleotide variants to a phenotypic trait, can have effects comparable to monogenic mutations. This review focuses on the emerging use of 'genome-wide' polygenic scores for plasma lipoproteins to define the etiology of clinical dyslipidemia, modify the severity of monogenic disease, and inform therapeutic options. RECENT FINDINGS Polygenic scores for low-density lipoprotein cholesterol (LDL-C), triglycerides, and high-density lipoprotein cholesterol are associated with severe hypercholesterolemia, hypertriglyceridemia, or hypoalphalipoproteinemia, respectively. These polygenic scores for LDL-C or triglycerides associate with risk of incident coronary artery disease (CAD) independent of polygenic scores designed specifically for CAD and may identify individuals that benefit most from lipid-lowering medication. Additionally, the severity of hypercholesterolemia and CAD associated with familial hypercholesterolemia-a common monogenic disorder-is modified by these polygenic factors. The current focus of polygenic scores for dyslipidemia is to design predictive polygenic scores for diverse populations and determining how these polygenic scores could be implemented and standardized for use in the clinic. SUMMARY Polygenic scores have shown early promise for the management of dyslipidemias, but several challenges need to be addressed before widespread clinical implementation to ensure that potential benefits are robust and reproducible, equitable, and cost-effective.
Collapse
Affiliation(s)
- Mark Trinder
- Centre for Heart Lung Innovation, University of British Columbia
- Experimental Medicine Program, University of British Columbia
| | - Liam R Brunham
- Centre for Heart Lung Innovation, University of British Columbia
- Experimental Medicine Program, University of British Columbia
- Department of Medicine, University of British Columbia
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
32
|
Ekoru K, Adeyemo AA, Chen G, Doumatey AP, Zhou J, Bentley AR, Shriner D, Rotimi CN. Genetic risk scores for cardiometabolic traits in sub-Saharan African populations. Int J Epidemiol 2021; 50:1283-1296. [PMID: 33729508 DOI: 10.1093/ije/dyab046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/25/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND There is growing support for the use of genetic risk scores (GRS) in routine clinical settings. Due to the limited diversity of current genomic discovery samples, there are concerns that the predictive power of GRS will be limited in non-European ancestry populations. GRS for cardiometabolic traits were evaluated in sub-Saharan Africans in comparison with African Americans and European Americans. METHODS We evaluated the predictive utility of GRS for 12 cardiometabolic traits in sub-Saharan Africans (AF; n = 5200), African Americans (AA; n = 9139) and European Americans (EUR; n = 9594). GRS were constructed as weighted sums of the number of risk alleles. Predictive utility was assessed using the additional phenotypic variance explained and the increase in discriminatory ability over traditional risk factors [age, sex and body mass index (BMI)], with adjustment for ancestry-derived principal components. RESULTS Across all traits, GRS showed up to a 5-fold and 20-fold greater predictive utility in EUR relative to AA and AF, respectively. Predictive utility was most consistent for lipid traits, with percentage increase in explained variation attributable to GRS ranging from 10.6% to 127.1% among EUR, 26.6% to 65.8% among AA and 2.4% to 37.5% among AF. These differences were recapitulated in the discriminatory power, whereby the predictive utility of GRS was 4-fold greater in EUR relative to AA and up to 44-fold greater in EUR relative to AF. Obesity and blood pressure traits showed a similar pattern of greater predictive utility among EUR. CONCLUSIONS This work demonstrates the poorer performance of GRS in AF and highlights the need to improve representation of multiple ethnic populations in genomic studies to ensure equitable clinical translation of GRS.
Collapse
Affiliation(s)
- Kenneth Ekoru
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Adebowale A Adeyemo
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ayo P Doumatey
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jie Zhou
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Amy R Bentley
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Shriner
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Charles N Rotimi
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
33
|
Niu M, Zhang L, Wang Y, Tu R, Liu X, Hou J, Huo W, Mao Z, Wang Z, Wang C. Genetic factors increase the identification efficiency of predictive models for dyslipidaemia: a prospective cohort study. Lipids Health Dis 2021; 20:11. [PMID: 33579296 PMCID: PMC7881493 DOI: 10.1186/s12944-021-01439-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 01/27/2021] [Indexed: 11/10/2022] Open
Abstract
Background Few studies have developed risk models for dyslipidaemia, especially for rural populations. Furthermore, the performance of genetic factors in predicting dyslipidaemia has not been explored. The purpose of this study is to develop and evaluate prediction models with and without genetic factors for dyslipidaemia in rural populations. Methods A total of 3596 individuals from the Henan Rural Cohort Study were included in this study. According to the ratio of 7:3, all individuals were divided into a training set and a testing set. The conventional models and conventional+GRS (genetic risk score) models were developed with Cox regression, artificial neural network (ANN), random forest (RF), and gradient boosting machine (GBM) classifiers in the training set. The area under the receiver operating characteristic curve (AUC), net reclassification index (NRI), and integrated discrimination index (IDI) were used to assess the discrimination ability of the models, and the calibration curve was used to show calibration ability in the testing set. Results Compared to the lowest quartile of GRS, the hazard ratio (HR) (95% confidence interval (CI)) of individuals in the highest quartile of GRS was 1.23(1.07, 1.41) in the total population. Age, family history of diabetes, physical activity, body mass index (BMI), triglycerides (TGs), high-density lipoprotein cholesterol (HDL-C), and low-density lipoprotein cholesterol (LDL-C) were used to develop the conventional models, and the AUCs of the Cox, ANN, RF, and GBM classifiers were 0.702(0.673, 0.729), 0.736(0.708, 0.762), 0.787 (0.762, 0.811), and 0.816(0.792, 0.839), respectively. After adding GRS, the AUCs increased by 0.005, 0.018, 0.023, and 0.015 with the Cox, ANN, RF, and GBM classifiers, respectively. The corresponding NRI and IDI were 25.6, 7.8, 14.1, and 18.1% and 2.3, 1.0, 2.5, and 1.8%, respectively. Conclusion Genetic factors could improve the predictive ability of the dyslipidaemia risk model, suggesting that genetic information could be provided as a potential predictor to screen for clinical dyslipidaemia. Trial registration The Henan Rural Cohort Study has been registered at the Chinese Clinical Trial Register. (Trial registration: ChiCTR-OOC-15006699. Registered 6 July 2015 - Retrospectively registered). Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01439-3.
Collapse
Affiliation(s)
- Miaomiao Niu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Liying Zhang
- School of Information Engineering, Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Yikang Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Runqi Tu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Xiaotian Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Jian Hou
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Wenqian Huo
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Zhenxing Mao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China
| | - Zhenfei Wang
- School of Information Engineering, Zhengzhou University, Zhengzhou, Henan, People's Republic of China.
| | - Chongjian Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, 450001, Henan, People's Republic of China.
| |
Collapse
|
34
|
Semaev S, Shakhtshneider E. Genetic Risk Score for Coronary Heart Disease: Review. J Pers Med 2020; 10:jpm10040239. [PMID: 33233501 PMCID: PMC7712936 DOI: 10.3390/jpm10040239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/05/2020] [Accepted: 11/17/2020] [Indexed: 12/27/2022] Open
Abstract
The present review deals with the stages of creation, methods of calculation, and the use of a genetic risk score for coronary heart disease in various populations. The concept of risk factors is generally recognized on the basis of the results of epidemiological studies in the 20th century; according to this concept, the high prevalence of diseases of the circulatory system is due to lifestyle characteristics and associated risk factors. An important and relevant task for the healthcare system is to identify the population segments most susceptible to cardiovascular diseases (CVDs). The level of individual risk of an unfavorable cardiovascular prognosis is determined by genetic factors in addition to lifestyle factors.
Collapse
Affiliation(s)
- Sergey Semaev
- Institute of Internal and Preventive Medicine—Branch of Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Bogatkova Str. 175/1, Novosibirsk 630089, Russia;
- Federal Research Center Institute of Cytology and Genetics, SB RAS, Prospekt Lavrentyeva 10, Novosibirsk 630090, Russia
| | - Elena Shakhtshneider
- Institute of Internal and Preventive Medicine—Branch of Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (SB RAS), Bogatkova Str. 175/1, Novosibirsk 630089, Russia;
- Federal Research Center Institute of Cytology and Genetics, SB RAS, Prospekt Lavrentyeva 10, Novosibirsk 630090, Russia
- Correspondence: or ; Tel./Fax: +7-(383)-264-2516
| |
Collapse
|
35
|
Vrablik M, Tichý L, Freiberger T, Blaha V, Satny M, Hubacek JA. Genetics of Familial Hypercholesterolemia: New Insights. Front Genet 2020; 11:574474. [PMID: 33133164 PMCID: PMC7575810 DOI: 10.3389/fgene.2020.574474] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/09/2020] [Indexed: 12/11/2022] Open
Abstract
Familial hypercholesterolemia (FH) is one of the most common monogenic diseases, leading to an increased risk of premature atherosclerosis and its cardiovascular complications due to its effect on plasma cholesterol levels. Variants of three genes (LDL-R, APOB and PCSK9) are the major causes of FH, but in some probands, the FH phenotype is associated with variants of other genes. Alternatively, the typical clinical picture of FH can result from the accumulation of common cholesterol-increasing alleles (polygenic FH). Although the Czech Republic is one of the most successful countries with respect to FH detection, approximately 80% of FH patients remain undiagnosed. The opportunities for international collaboration and experience sharing within international programs (e.g., EAS FHSC, ScreenPro FH, etc.) will improve the detection of FH patients in the future and enable even more accessible and accurate genetic diagnostics.
Collapse
Affiliation(s)
- Michal Vrablik
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia
| | - Lukas Tichý
- Centre of Molecular Biology and Gene Therapy, University Hospital, Brno, Czechia
| | - Tomas Freiberger
- Centre for Cardiovascular Surgery and Transplantation, Brno, and Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Vladimir Blaha
- Internal Gerontometabolic Department, Charles University and University Hospital Hradec Kralove, Hradec Kralove, Czechia
| | - Martin Satny
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia
| | - Jaroslav A Hubacek
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czechia.,Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, Prague, Czechia
| |
Collapse
|
36
|
Polygenic Markers in Patients Diagnosed of Autosomal Dominant Hypercholesterolemia in Catalonia: Distribution of Weighted LDL-c-Raising SNP Scores and Refinement of Variant Selection. Biomedicines 2020; 8:biomedicines8090353. [PMID: 32942679 PMCID: PMC7554998 DOI: 10.3390/biomedicines8090353] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/03/2020] [Accepted: 09/10/2020] [Indexed: 11/16/2022] Open
Abstract
Familial hypercholesterolemia (FH) is associated with mutations in the low-density lipoprotein (LDL) receptor (LDLR), apolipoprotein B (APOB), and proprotein convertase subtilisin/kexin 9 (PCSK9) genes. A pathological variant has not been identified in 30-70% of clinically diagnosed FH patients, and a burden of LDL cholesterol (LDL-c)-raising alleles has been hypothesized as a potential cause of hypercholesterolemia in these patients. Our aim was to study the distribution of weighted LDL-c-raising single-nucleotide polymorphism (SNP) scores (weighted gene scores or wGS) in a population recruited in a clinical setting in Catalonia. The study included 670 consecutive patients with a clinical diagnosis of FH and a prior genetic study involving 250 mutation-positive (FH/M+) and 420 mutation-negative (FH/M-) patients. Three wGSs based on LDL-c-raising variants were calculated to evaluate their distribution among FH patients and compared with 503 European samples from the 1000 Genomes Project. The FH/M- patients had significantly higher wGSs than the FH/M+ and control populations, with sensitivities ranging from 42% to 47%. A wGS based only on the SNPs significantly associated with FH (wGS8) showed a higher area under the receiver operating characteristic curve, and higher diagnostic specificity and sensitivity, with 46.4% of the subjects in the top quartile. wGS8 would allow for the assignment of a genetic cause to 66.4% of the patients if those with polygenic FH are added to the 37.3% of patients with monogenic FH. Our data indicate that a score based on 8 SNPs and the75th percentile cutoff point may identify patients with polygenic FH in Catalonia, although with limited diagnostic sensitivity and specificity.
Collapse
|
37
|
Rieck L, Bardey F, Grenkowitz T, Bertram L, Helmuth J, Mischung C, Spranger J, Steinhagen-Thiessen E, Bobbert T, Kassner U, Demuth I. Mutation spectrum and polygenic score in German patients with familial hypercholesterolemia. Clin Genet 2020; 98:457-467. [PMID: 32770674 DOI: 10.1111/cge.13826] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/17/2022]
Abstract
Autosomal-dominant familial hypercholesterolemia (FH) is characterized by increased plasma concentrations of low-density lipoprotein cholesterol (LDL-C) and a substantial risk to develop cardiovascular disease. Causative mutations in three major genes are known: the LDL receptor gene (LDLR), the apolipoprotein B gene (APOB) and the proprotein convertase subtilisin/kexin 9 gene (PCSK9). We clinically characterized 336 patients suspected to have FH and screened them for disease causing mutations in LDLR, APOB, and PCSK9. We genotyped six single nucleotide polymorphisms (SNPs) to calculate a polygenic risk score for the patients and 1985 controls. The 117 patients had a causative variant in one of the analyzed genes. Most variants were found in the LDLR gene (84.9%) with 11 novel mutations. The mean polygenic risk score was significantly higher in FH mutation negative subjects than in FH mutation positive patients (P < .05) and healthy controls (P < .001), whereas the score of the two latter groups did not differ significantly. However, the score explained only about 3% of the baseline LDL-C variance. We verified the previously described clinical and genetic variability of FH for German hypercholesterolemic patients. Evaluation of a six-SNP polygenic score recently proposed for clinical use suggests that it is not a reliable tool to classify hypercholesterolemic patients.
Collapse
Affiliation(s)
- Lorenz Rieck
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Frieda Bardey
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Grenkowitz
- Department of Cardiology, Charité - University Medicine Berlin (Campus Benjamin Franklin), Berlin, Germany
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany.,Center for Lifespan Changes in Brain and Cognition (LCBC), Dept of Psychology, University of Oslo, Oslo, Norway
| | - Johannes Helmuth
- Department Molecular Diagnostics, Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Claudia Mischung
- Department Molecular Diagnostics, Labor Berlin - Charité Vivantes GmbH, Berlin, Germany
| | - Joachim Spranger
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Elisabeth Steinhagen-Thiessen
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Bobbert
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ursula Kassner
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ilja Demuth
- Department of Endocrinology and Metabolism, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Charité - Universitätsmedizin Berlin, BCRT - Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
38
|
Paquette M, Fantino M, Bernard S, Baass A. The ZPR1 genotype predicts myocardial infarction in patients with familial hypercholesterolemia. J Clin Lipidol 2020; 14:660-666. [DOI: 10.1016/j.jacl.2020.07.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
|
39
|
Trinder M, Paquette M, Cermakova L, Ban MR, Hegele RA, Baass A, Brunham LR. Polygenic Contribution to Low-Density Lipoprotein Cholesterol Levels and Cardiovascular Risk in Monogenic Familial Hypercholesterolemia. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2020; 13:515-523. [PMID: 33079599 PMCID: PMC7889287 DOI: 10.1161/circgen.120.002919] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is a common autosomal codominant genetic disorder, which causes elevated levels of low-density lipoprotein cholesterol (LDL-C) and increased risk of premature atherosclerotic cardiovascular disease (ASCVD). Even among individuals with monogenic FH, there is substantial interindividual variability in LDL-C levels and risk of ASCVD. We assessed the influence of an LDL-C polygenic score on levels of LDL-C and risk of ASCVD for individuals with monogenic FH. METHODS We constructed a weighted LDL-C polygenic score, composed of 28 single-nucleotide variants, for individuals with monogenic FH from the British Columbia FH (n=262); Nutrition, Metabolism and Atherosclerosis Clinic (n=552); and UK Biobank cohorts (n=306). We assessed the association between LDL-C polygenic score with LDL-C levels and ASCVD risk using linear regression and Cox-proportional hazard models, respectively. ASCVD was defined as myocardial infarction, coronary or carotid revascularization, transient ischemic attack, or stroke. The results from individual cohorts were combined in fixed-effect meta-analyses. RESULTS Levels of LDL-C were significantly associated with LDL-C polygenic score in the Nutrition, Metabolism and Atherosclerosis Clinic cohort, UK Biobank cohort, and in the meta-analysis (β [95% CI]=0.13 [0.072-0.19] per a 20% increase in LDL-C polygenic score percentile, P<0.0001). Additionally, an elevated LDL-C polygenic score (≥80th percentile) was associated with a trend towards increased ASCVD risk in all 3 cohorts individually. This association was statistically significant in the meta-analysis (hazard ratio [95% CI]=1.48 [1.02-2.14], P=0.04). CONCLUSIONS Polygenic contributions to LDL-C explain some of the heterogeneity in clinical presentation and ASCVD risk for individuals with FH.
Collapse
Affiliation(s)
- Mark Trinder
- Centre for Heart Lung Innovation (M.T., L.R.B.), University of British Columbia, Vancouver.,Experimental Medicine Program (M.T., L.R.B.), University of British Columbia, Vancouver
| | - Martine Paquette
- Nutrition, Metabolism and Atherosclerosis Clinic, Institut de recherches cliniques de Montréal, Quebec (M.P., A.B.)
| | - Lubomira Cermakova
- Healthy Heart Program Prevention Clinic, St Paul's Hospital, Vancouver, British Columbia (L.C., L.R.B.)
| | - Matthew R Ban
- Departments of Medicine and Biochemistry, Schulich School of Medicine and Dentistry and Robarts Research Institute, Western University, London, ON (M.R.B., R.A.H.)
| | - Robert A Hegele
- Departments of Medicine and Biochemistry, Schulich School of Medicine and Dentistry and Robarts Research Institute, Western University, London, ON (M.R.B., R.A.H.)
| | - Alexis Baass
- Nutrition, Metabolism and Atherosclerosis Clinic, Institut de recherches cliniques de Montréal, Quebec (M.P., A.B.).,Departments of Medicine, McGill University, Montreal, Quebec, Canada (A.B.)
| | - Liam R Brunham
- Centre for Heart Lung Innovation (M.T., L.R.B.), University of British Columbia, Vancouver.,Experimental Medicine Program (M.T., L.R.B.), University of British Columbia, Vancouver.,Departments of Medicine and Medical Genetics (L.R.B.), University of British Columbia, Vancouver.,Healthy Heart Program Prevention Clinic, St Paul's Hospital, Vancouver, British Columbia (L.C., L.R.B.)
| |
Collapse
|
40
|
Turkowski KL, Dotzler SM, Tester DJ, Giudicessi JR, Bos JM, Speziale AD, Vollenweider JM, Ackerman MJ. Corrected QT Interval–Polygenic Risk Score and Its Contribution to Type 1, Type 2, and Type 3 Long-QT Syndrome in Probands and Genotype-Positive Family Members. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2020; 13:e002922. [DOI: 10.1161/circgen.120.002922] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background:
Long-QT syndrome (LQTS) is characterized by a prolonged heart rate–corrected QT interval (QTc). Genome-wide association studies identified common genetic variants that collectively explain ≈8% to 10% of QTc variation in the general population.
Methods:
Overall, 423 patients with LQT1, LQT2, or LQT3 were genotyped for 61 QTc-associated genetic variants used in a prototype QTc–polygenic risk score (QTc-PRS). A weighted QTc-PRS (range, 0–154.8 ms) was calculated for each patient, and the FHS (Framingham Heart Study) population-based reference cohort (n=853).
Results:
The average QTc-PRS in LQTS was 88.0±7.2 and explained only ≈2.0% of the QTc variability. The QTc-PRS in LQTS probands (n=137; 89.3±6.8) was significantly greater than both FHS controls (87.2±7.4, difference-in-means±SE: 2.1±0.7,
P
<0.002) and LQTS genotype-positive family members (87.5±7.4, difference-in-mean, 1.8±.7,
P
<0.009). There was no difference in QTc-PRS between symptomatic (n=156, 88.6±7.3) and asymptomatic patients (n=267; 87.7±7.2, difference-in-mean, 0.9±0.7, P=0.15). LQTS patients with a QTc≥480 ms (n=120) had a significantly higher QTc-PRS (89.3±6.7) than patients with a QTc<480 ms (n=303, 87.6±7.4, difference-in-mean, 1.7±0.8,
P
<0.05). There was no difference in QTc-PRS or QTc between genotypes.
Conclusions:
The QTc-PRS explained <2% of the QTc variability in our LQT1, LQT2, and LQT3 cohort, contributing 5× less to their QTc value than in the general population. This prototype QTc-PRS does not distinguish/predict the clinical outcomes of individuals with LQTS.
Collapse
Affiliation(s)
- Kari L. Turkowski
- Mayo Clinic Graduate School of Biomedical Sciences (K.L.T., S.M.D.), Mayo Clinic, Rochester, MN, USA
- Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology & Experimental Therapeutics (K.L.T., S.M.D., D.J.T., J.M.B., M.J.A.), Mayo Clinic, Rochester, MN, USA
| | - Steven M. Dotzler
- Mayo Clinic Graduate School of Biomedical Sciences (K.L.T., S.M.D.), Mayo Clinic, Rochester, MN, USA
- Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology & Experimental Therapeutics (K.L.T., S.M.D., D.J.T., J.M.B., M.J.A.), Mayo Clinic, Rochester, MN, USA
| | - David J. Tester
- Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology & Experimental Therapeutics (K.L.T., S.M.D., D.J.T., J.M.B., M.J.A.), Mayo Clinic, Rochester, MN, USA
| | - John R. Giudicessi
- Clinician-Investigator Training Program, Department of Cardiovascular Medicine (J.R.G.), Mayo Clinic, Rochester, MN, USA
| | - J. Martijn Bos
- Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology & Experimental Therapeutics (K.L.T., S.M.D., D.J.T., J.M.B., M.J.A.), Mayo Clinic, Rochester, MN, USA
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine (J.M.B., M.J.A.), Mayo Clinic, Rochester, MN, USA
| | - Ashley D. Speziale
- Medical Genome Facility (A.D.S., J.M.V.), Mayo Clinic, Rochester, MN, USA
| | | | - Michael J. Ackerman
- Windland Smith Rice Sudden Death Genomics Laboratory, Department of Molecular Pharmacology & Experimental Therapeutics (K.L.T., S.M.D., D.J.T., J.M.B., M.J.A.), Mayo Clinic, Rochester, MN, USA
- Division of Heart Rhythm Services, Windland Smith Rice Genetic Heart Rhythm Clinic, Department of Cardiovascular Medicine (M.J.A.), Mayo Clinic, Rochester, MN, USA
- Division of Pediatric Cardiology, Department of Pediatric and Adolescent Medicine (J.M.B., M.J.A.), Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
41
|
Mahtta D, Khalid U, Misra A, Samad Z, Nasir K, Virani SS. Premature Atherosclerotic Cardiovascular Disease: What Have We Learned Recently? Curr Atheroscler Rep 2020; 22:44. [PMID: 32671484 DOI: 10.1007/s11883-020-00862-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW In contrast to patients with non-premature atherosclerotic cardiovascular disease (ASCVD), patients with premature ASCVD have not observed a similar decline in cardiovascular mortality and recurrent adverse events. We sought to review the underlying risk factors, potential gaps in medical management, associated outcomes, and tools for risk prognostication among patients with premature ASCVD. RECENT FINDINGS In addition to traditional cardiovascular risk factors (i.e., diabetes, familial hypercholesterolemia), non-traditional risk factors such as chronic inflammatory conditions, recreational drug use, genetics, and pregnancy-related complications play a key role in development and progression of premature ASCVD. Patients with premature ASCVD, and especially women, receive less optimal medical management as compared to their non-premature counterparts. There is an increasing prevalence of cardiovascular risk factors among young adults. Hence, this population remains at an elevated risk for premature ASCVD and subsequent adverse cardiovascular events. Future studies evaluating different risk assessment tools and focusing on young patients across all three major domains of ASCVD are needed.
Collapse
Affiliation(s)
- Dhruv Mahtta
- Health Policy, Quality & Informatics Program, Michael E. DeBakey VA Medical Center Health Services Research & Development Center for Innovations in Quality, Effectiveness, and Safety, Houston, TX, USA.,Department of Medicine, Section of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Umair Khalid
- Department of Medicine, Section of Cardiology, Baylor College of Medicine, Houston, TX, USA.,Section of Cardiology, Health Services Research and Development (152), Michael E. DeBakey Veterans Affairs Medical Center, 2002 Holcombe Blvd., Houston, TX, 77030, USA
| | - Arunima Misra
- Department of Medicine, Section of Cardiology, Baylor College of Medicine, Houston, TX, USA.,Section of Cardiology, Health Services Research and Development (152), Michael E. DeBakey Veterans Affairs Medical Center, 2002 Holcombe Blvd., Houston, TX, 77030, USA
| | - Zainab Samad
- Department of Medicine, The Aga Khan University, Karachi, Pakistan
| | - Khurram Nasir
- Methodist DeBakey Heart & Vascular Center, Houston Methodist Hospital, Houston, TX, USA
| | - Salim S Virani
- Health Policy, Quality & Informatics Program, Michael E. DeBakey VA Medical Center Health Services Research & Development Center for Innovations in Quality, Effectiveness, and Safety, Houston, TX, USA. .,Department of Medicine, Section of Cardiology, Baylor College of Medicine, Houston, TX, USA. .,Section of Cardiology, Health Services Research and Development (152), Michael E. DeBakey Veterans Affairs Medical Center, 2002 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
42
|
Yanes T, McInerney-Leo AM, Law MH, Cummings S. The emerging field of polygenic risk scores and perspective for use in clinical care. Hum Mol Genet 2020; 29:R165-R176. [DOI: 10.1093/hmg/ddaa136] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Abstract
Abstract
Genetic testing is used widely for diagnostic, carrier and predictive testing in monogenic diseases. Until recently, there were no genetic testing options available for multifactorial complex diseases like heart disease, diabetes and cancer. Genome-wide association studies (GWAS) have been invaluable in identifying single-nucleotide polymorphisms (SNPs) associated with increased or decreased risk for hundreds of complex disorders. For a given disease, SNPs can be combined to generate a cumulative estimation of risk known as a polygenic risk score (PRS). After years of research, PRSs are increasingly used in clinical settings. In this article, we will review the literature on how both genome-wide and restricted PRSs are developed and the relative merit of each. The validation and evaluation of PRSs will also be discussed, including the recognition that PRS validity is intrinsically linked to the methodological and analytical approach of the foundation GWAS together with the ethnic characteristics of that cohort. Specifically, population differences may affect imputation accuracy, risk magnitude and direction. Even as PRSs are being introduced into clinical practice, there is a push to combine them with clinical and demographic risk factors to develop a holistic disease risk. The existing evidence regarding the clinical utility of PRSs is considered across four different domains: informing population screening programs, guiding therapeutic interventions, refining risk for families at high risk, and facilitating diagnosis and predicting prognostic outcomes. The evidence for clinical utility in relation to five well-studied disorders is summarized. The potential ethical, legal and social implications are also highlighted.
Collapse
Affiliation(s)
- Tatiane Yanes
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Aideen M McInerney-Leo
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Matthew H Law
- Statistical Genetics Lab, QIMR Berghofer Medical Research Institute, Herston QLD 4006, Australia
- Faculty of Health, School of Biomedical Sciences, and Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove QLD 4059, Australia
| | | |
Collapse
|
43
|
Association Study of Coronary Artery Disease-Associated Genome-Wide Significant SNPs with Coronary Stenosis in Pakistani Population. DISEASE MARKERS 2020; 2020:9738567. [PMID: 32685059 PMCID: PMC7336215 DOI: 10.1155/2020/9738567] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 01/17/2020] [Accepted: 01/23/2020] [Indexed: 11/19/2022]
Abstract
Genome-wide association studies (GWAS) of coronary artery disease (CAD) have revealed multiple genetic risk loci. We assessed the association of 47 genome-wide significant single-nucleotide polymorphisms (SNPs) at 43 CAD loci with coronary stenosis in a Pakistani sample comprising 663 clinically ascertained and angiographically confirmed cases. Genotypes were determined using the iPLEX Gold technology. All statistical analyses were performed using R software. Linkage disequilibrium (LD) between significant SNPs was determined using SNAP web portal, and functional annotation of SNPs was performed using the RegulomeDB and Genotype-Tissue Expression (GTEx) databases. Genotyping comparison was made between cases with severe stenosis (≥70%) and mild/minimal stenosis (<30%). Five SNPs demonstrated significant associations: three with additive genetic models PLG/rs4252120 (p = 0.0078), KIAA1462/rs2505083 (p = 0.005), and SLC22A3/rs2048327 (p = 0.045) and two with recessive models SORT1/rs602633 (p = 0.005) and UBE2Z/rs46522 (p = 0.03). PLG/rs4252120 was in LD with two functional PLG variants (rs4252126 and rs4252135), each with a RegulomeDB score of 1f. Likewise, KIAA1462/rs2505083 was in LD with a functional SNP, KIAA1462/rs3739998, having a RegulomeDB score of 2b. In the GTEx database, KIAA1462/rs2505083, SLC22A3/rs2048327, SORT1/rs602633, and UBE2Z/rs46522 SNPs were found to be expression quantitative trait loci (eQTLs) in CAD-associated tissues. In conclusion, five genome-wide significant SNPs previously reported in European GWAS were replicated in the Pakistani sample. Further association studies on larger non-European populations are needed to understand the worldwide genetic architecture of CAD.
Collapse
|
44
|
Loika Y, Irincheeva I, Culminskaya I, Nazarian A, Kulminski AM. Polygenic risk scores: pleiotropy and the effect of environment. GeroScience 2020; 42:1635-1647. [PMID: 32488673 DOI: 10.1007/s11357-020-00203-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/08/2020] [Indexed: 10/24/2022] Open
Abstract
Polygenic risk scores (PRSs) discriminate trait risks better than single genetic markers because they aggregate the effects of risk alleles from multiple genetic loci. Constructing pleiotropic PRSs and understanding heterogeneity, and the replication of PRS-trait associations can strengthen its applications. By using variational Bayesian multivariate high-dimensional regression, we constructed pleiotropic PRSs jointly associated with body mass index, systolic and diastolic blood pressure, total and high-density lipoprotein cholesterol in a sample of 18,108 Caucasians from three independent cohorts. We found that dissecting heterogeneity associated with birth year, which is a proxy of exogenous exposures, improved the replication of significant PRS-trait associations from 37.5% (6 of 16) in the entire sample to 90% (18 of 20) in the more homogeneous sample of individuals born before the year 1925. Our findings suggest that secular changes in exogenous exposures may substantially modify pleiotropic risk profiles affecting translation of genetic discoveries into health care.
Collapse
Affiliation(s)
- Yury Loika
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27708-0408, USA.
| | - Irina Irincheeva
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27708-0408, USA.
| | - Irina Culminskaya
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27708-0408, USA
| | - Alireza Nazarian
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27708-0408, USA
| | - Alexander M Kulminski
- Biodemography of Aging Research Unit, Social Science Research Institute, Duke University, Durham, NC, 27708-0408, USA.
| |
Collapse
|
45
|
Semenova AE, Sergienko IV, García-Giustiniani D, Monserrat L, Popova AB, Nozadze DN, Ezhov MV. Verification of Underlying Genetic Cause in a Cohort of Russian Patients with Familial Hypercholesterolemia Using Targeted Next Generation Sequencing. J Cardiovasc Dev Dis 2020; 7:jcdd7020016. [PMID: 32423031 PMCID: PMC7345545 DOI: 10.3390/jcdd7020016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/28/2020] [Accepted: 05/08/2020] [Indexed: 01/25/2023] Open
Abstract
Russian patients with familial hypercholesterolemia (FH) were screened for pathogenic mutations using targeted next generation sequencing. Genetic testing was performed in 52 probands with definite or probable FH based on the Dutch lipid clinic network criteria (DLCN score ≥6). Blood samples were studied by massive parallel sequencing (Illumina HiSeq 1500 platform) using a custom capture library related to dyslipidemia and premature atherosclerosis. Mutations considered to be responsible for monogenic FH were identified in 48% of the probands: 24 with mutations in the LDLR gene and two with a mutation in the APOB gene. There were 22 pathogenic/likely pathogenic mutations in LDLR, eight of which have not been previously described in the literature. Four patients with a clinical picture of homozygous FH had two heterozygous LDLR mutations. Although mutation-negative patients had highly elevated total cholesterol and low-density lipoprotein cholesterol levels, only half of them had a family history of hypercholesterolemia. With respect to heterozygous FH, mutation-positive patients had higher maximum total cholesterol levels (p = 0.01), more severe carotid atherosclerotic lesions, and a higher percentage of premature peripheral artery disease (p = 0.03) than mutation-negative ones. However, the number of patients who suffered from myocardial infarction was similar between the two groups.
Collapse
Affiliation(s)
- Anna E. Semenova
- National Medical Research Center of Cardiology, 121552 Moscow, Russia; (I.V.S.); (A.B.P.); (D.N.N.); (M.V.E.)
- Health in Code SL, Clinical Department, 15006 A Coruña, Spain; (D.G.-G.); (L.M.)
- Correspondence: ; Tel.: +7-926-239-4171
| | - Igor V. Sergienko
- National Medical Research Center of Cardiology, 121552 Moscow, Russia; (I.V.S.); (A.B.P.); (D.N.N.); (M.V.E.)
| | | | - Lorenzo Monserrat
- Health in Code SL, Clinical Department, 15006 A Coruña, Spain; (D.G.-G.); (L.M.)
| | - Anna B. Popova
- National Medical Research Center of Cardiology, 121552 Moscow, Russia; (I.V.S.); (A.B.P.); (D.N.N.); (M.V.E.)
| | - Diana N. Nozadze
- National Medical Research Center of Cardiology, 121552 Moscow, Russia; (I.V.S.); (A.B.P.); (D.N.N.); (M.V.E.)
| | - Marat V. Ezhov
- National Medical Research Center of Cardiology, 121552 Moscow, Russia; (I.V.S.); (A.B.P.); (D.N.N.); (M.V.E.)
| |
Collapse
|
46
|
Brunham LR, Mancini GBJ. Editorial Commentary: What Determines the Risk of Cardiovascular Disease in Familial Hypercholesterolemia? Trends Cardiovasc Med 2020; 31:216-217. [PMID: 32407995 DOI: 10.1016/j.tcm.2020.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 11/27/2022]
Affiliation(s)
- Liam R Brunham
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
| | - G B John Mancini
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
47
|
Rader DJ, Sheth S. Polygenic Risk Scores in Familial Hypercholesterolemia. J Am Coll Cardiol 2020; 74:523-525. [PMID: 31345426 DOI: 10.1016/j.jacc.2019.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Daniel J Rader
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Samip Sheth
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
48
|
Aragam KG, Natarajan P. Polygenic Scores to Assess Atherosclerotic Cardiovascular Disease Risk: Clinical Perspectives and Basic Implications. Circ Res 2020; 126:1159-1177. [PMID: 32324503 PMCID: PMC7926201 DOI: 10.1161/circresaha.120.315928] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
An individual's susceptibility to atherosclerotic cardiovascular disease is influenced by numerous clinical and lifestyle factors, motivating the multifaceted approaches currently endorsed for primary and secondary cardiovascular disease prevention. With growing knowledge of the genetic basis of atherosclerotic cardiovascular disease-in particular, coronary artery disease-and its contribution to disease pathogenesis, there is increased interest in understanding the potential clinical utility of a genetic predictor that might further refine the assessment and management of atherosclerotic cardiovascular disease risk. Rapid scientific and technological advances have enabled widespread genotyping efforts and dynamic research in the field of coronary artery disease genetic risk prediction. In this review, we describe how genomic analyses of coronary artery disease have been leveraged to create polygenic risk scores. We then discuss evaluations of the clinical utility of these scores, pertinent mechanistic insights gleaned, and practical considerations relevant to the implementation of polygenic risk scores in the health care setting.
Collapse
Affiliation(s)
- Krishna G. Aragam
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Pradeep Natarajan
- Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
49
|
Bianconi V, Banach M, Pirro M. Why patients with familial hypercholesterolemia are at high cardiovascular risk? Beyond LDL-C levels. Trends Cardiovasc Med 2020; 31:205-215. [PMID: 32205033 DOI: 10.1016/j.tcm.2020.03.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 02/03/2020] [Accepted: 03/06/2020] [Indexed: 01/07/2023]
Abstract
Familial hypercholesterolemia (FH) is a common genetic cause of elevated low-density lipoprotein cholesterol (LDL-C) due to defective clearance of circulating LDL particles. All FH patients are at high risk for premature cardiovascular disease (CVD) events due to their genetically determined lifelong exposure to high LDL-C levels. However, different rates of CVD events have been reported in FH patients, even among those with the same genetic mutations and comparable LDL-C levels. Hence, additional CVD risk modifiers, beyond LDL-C, may contribute to increase CVD risk in the FH population. In this review, we discuss the overall CVD risk burden of the FH population. Additionally, we revise the prognostic impact of several traditional and emerging predictors of CVD risk and we provide an overview of the role of specific tools to stratify CVD risk in FH patients in order to ensure them a more personalized treatment approach.
Collapse
Affiliation(s)
- Vanessa Bianconi
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Hospital "Santa Maria della Misericordia", Piazzale Menghini, 1, 06129 Perugia, Italy
| | - Maciej Banach
- Department of Hypertension, Chair of Nephrology and Hypertension, WAM University Hospital in Lodz, Medical University of Lodz, Zeromskiego 113, 90-549 Lodz, Poland; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland.
| | - Matteo Pirro
- Unit of Internal Medicine, Department of Medicine, University of Perugia, Hospital "Santa Maria della Misericordia", Piazzale Menghini, 1, 06129 Perugia, Italy.
| | | |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Extensive work has gone into understanding the genetics of cardiovascular disease (CVD) and implicating genes involved in hyperlipidaemia. Translation into routine practise involves using genetic risk scores (GRS) to identify high-risk individuals in the general population. Some of these risk scores are beginning to disentangle the complex nature of CVD and inherited dyslipidaemias. RECENT FINDINGS GRS of varying complexity have been used to identify high-risk groups of patients with polygenic CVD including some individuals with risk equivalent to monogenic disease. In phenotypic familial hypercholesterolaemia a six or 12 gene lipid GRS may identify polygenic cases that comprise up to 50% of cases. In high triglyceride syndromes including even cases of familial chylomicronaemia syndrome more than 80% of cases are polygenic and not even associated with rare variants. In both familial hypercholesterolaemia and familial chylomicronaemia syndrome individuals with polygenic disease have a lower risk than those with monogenic disease. SUMMARY GRS show promise in identifying individuals with high risks of CVD. They have a close relationship with imaging markers. It is unclear whether GRS, imaging or both will be used to identify individuals at high risk of future events.
Collapse
|