1
|
Schormair B. Genetics of Restless Legs Syndrome: Insights from Genome-Wide Association Studies. Sleep Med Clin 2025; 20:193-202. [PMID: 40348531 DOI: 10.1016/j.jsmc.2025.02.008] [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: 05/14/2025]
Abstract
Genome-wide association studies (GWAS) of common and low-frequency variants have discovered 164 genetic risk loci for restless legs syndrome (RLS) in adult populations of European ancestry. Sex-specific GWAS meta-analyses revealed largely overlapping genetic risk profiles for women and men and are in line with a nongenetic risk factor driving the higher prevalence seen in women. Genetic investigations of pediatric RLS are limited, but the likely inclusion of early-onset cases in GWAS of adult populations and the similar phenotypic presentation of both forms suggest that genetic risk variants identified in adult populations transfer to pediatric RLS.
Collapse
Affiliation(s)
- Barbara Schormair
- Institute of Neurogenomics, Computational Health Department, Helmholtz Zentrum München - German Research Center for Environmental Health (GmbH), Ingolstaedter Landstrasse 1, Neuherberg 85764, Germany; Institute of Human Genetics, TUM School of Medicine and Health, Technical University Munich, Munich, Germany.
| |
Collapse
|
2
|
Trotter PM, Merner AR, Ginn LA, Martinez AC, Battaglino AL, Craig KP, Bach J, Freedberg KJ, Soda T, Storch EA, Lázaro-Muñoz G, Pereira S. Navigating the Future of Polygenic Risk Scores: Insights from Child and Adolescent Psychiatrists. Child Psychiatry Hum Dev 2025:10.1007/s10578-025-01854-y. [PMID: 40381156 DOI: 10.1007/s10578-025-01854-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 05/19/2025]
Abstract
Polygenic risk scores (PRS) are a method of calculating genetic risk for polygenic, or multi-gene, disorders. These scores have potential impacts in the realm of child and adolescent psychiatry, given the high prevalence of psychiatric disorders among youth. However, there are concerns about PRS implementation among key stakeholders, namely child and adolescent psychiatrists (CAP). We conducted interviews with 29 U.S.-based CAP to investigate clinician attitudes toward the use of PRS. The data herein correspond to a future scenario we provided CAP in which PRS are accurate and portable to patients of different racial and ethnic backgrounds. We found that CAP envisioned some utility for PRS in regards to clinical surveillance and treatment; however, several desired further research demonstrating that PRS positively impact patient outcomes before they would consider implementing PRS in-clinic. The most cited concern about PRS deployment was the potential for misinterpretation and misuse of PRS on the part of patients, families, corporate entities, and clinicians themselves. CAP emphasized the need for support in the form of testing infrastructure, clinical guidelines, and collaboration with the involvement of PRS experts, when considering PRS applications in the future.
Collapse
Affiliation(s)
- Page M Trotter
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, One Baylor Plaza, Suite 310D, Houston, TX, 77030, USA
| | - Amanda R Merner
- Center for Bioethics, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Lauren A Ginn
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, One Baylor Plaza, Suite 310D, Houston, TX, 77030, USA
- Department of Biosciences, Rice University, Houston, TX, USA
| | - Abigail C Martinez
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, One Baylor Plaza, Suite 310D, Houston, TX, 77030, USA
| | | | | | - Jason Bach
- University of Pennsylvania Carey Law School, Philadelphia, PA, USA
| | | | - Takahiro Soda
- Department of Psychiatry, University of Florida, Gainesville, FL, USA
- Center for Autism and Neurodevelopment, University of Florida, Gainesville, FL, USA
| | - Eric A Storch
- Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Gabriel Lázaro-Muñoz
- Center for Bioethics, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA
| | - Stacey Pereira
- Center for Medical Ethics and Health Policy, Baylor College of Medicine, One Baylor Plaza, Suite 310D, Houston, TX, 77030, USA.
| |
Collapse
|
3
|
Solomon BD, Cheatham M, de Guimarães TAC, Duong D, Haendel MA, Hsieh TC, Javanmardi B, Johnson B, Krawitz P, Kruszka P, Laurent T, Lee NC, McWalter K, Michaelides M, Mohnike K, Pontikos N, Guillen Sacoto MJ, Shwetar YJ, Ustach VD, Waikel RL, Woof W. Perspectives on the Current and Future State of Artificial Intelligence in Medical Genetics. Am J Med Genet A 2025:e64118. [PMID: 40375359 DOI: 10.1002/ajmg.a.64118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/14/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
Artificial intelligence (AI) is rapidly transforming numerous aspects of daily life, including clinical practice and biomedical research. In light of this rapid transformation, and in the context of medical genetics, we assembled a group of leaders in the field to respond to the question about how AI is affecting, and especially how AI will affect, medical genetics. The authors who contributed to this collection of essays intentionally represent different areas of expertise, career stages, and geographies, and include diverse types of clinicians, computer scientists, and researchers. The individual pieces cover a wide range of areas related to medical genetics; we expect that these pieces may provide helpful windows into the ways in which AI is being actively studied, used, and considered in medical genetics.
Collapse
Affiliation(s)
- Benjamin D Solomon
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, Maryland, USA
| | - Morgan Cheatham
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Thales A C de Guimarães
- Moorfields Eye Hospital National Health Service Foundation Trust, London, UK
- University College London Institute of Ophthalmology, London, UK
- National Institute for Health and Care Research Moorfields Biomedical Research Centre, London, UK
| | - Dat Duong
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, Maryland, USA
| | - Melissa A Haendel
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Tzung-Chien Hsieh
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Behnam Javanmardi
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | | | | | - Ni-Chung Lee
- Department of Pediatrics and Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | | | - Michel Michaelides
- Moorfields Eye Hospital National Health Service Foundation Trust, London, UK
- University College London Institute of Ophthalmology, London, UK
- National Institute for Health and Care Research Moorfields Biomedical Research Centre, London, UK
| | - Klaus Mohnike
- Children's Hospital, Otto-von-Guericke-University, Magdeburg, Germany
| | - Nikolas Pontikos
- Moorfields Eye Hospital National Health Service Foundation Trust, London, UK
- University College London Institute of Ophthalmology, London, UK
- National Institute for Health and Care Research Moorfields Biomedical Research Centre, London, UK
| | | | - Yousif J Shwetar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | - Rebekah L Waikel
- Medical Genomics Unit, National Human Genome Research Institute, Bethesda, Maryland, USA
| | - William Woof
- University College London Institute of Ophthalmology, London, UK
- National Institute for Health and Care Research Moorfields Biomedical Research Centre, London, UK
| |
Collapse
|
4
|
Long E, Williams J, Zhang H, Choi J. An evolving understanding of multiple causal variants underlying genetic association signals. Am J Hum Genet 2025; 112:741-750. [PMID: 39965570 DOI: 10.1016/j.ajhg.2025.01.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/20/2025] Open
Abstract
Understanding how genetic variation contributes to phenotypic variation is a fundamental question in genetics. Genome-wide association studies (GWASs) have discovered numerous genetic associations with various human phenotypes, most of which contain co-inherited variants in strong linkage disequilibrium (LD) with indistinguishable statistical significance. The experimental and analytical difficulty in identifying the "causal variant" among the co-inherited variants has traditionally led mechanistic studies to focus on relatively simple loci, where a single functional variant is presumed to explain most of the association signal and affect a target gene. The notion that a single causal variant is responsible for an association signal, while other variants in LD are merely correlated, has often been assumed in functional studies. However, emerging evidence powered by high-throughput experimental tools and context-specific functional databases argues that even a single independent signal may involve multiple functional variants in strong LD, each contributing to the observed genetic association. In this perspective, we articulate this evolving understanding of causal variants through examples from both traditional locus-by-locus approaches and more recent high-throughput functional studies. We then discuss the implications and prospects of this notion in understanding the genetic architecture of complex traits and interpreting the variant-level causality in GWAS follow-up studies.
Collapse
Affiliation(s)
- Erping Long
- State Key Laboratory of Respiratory Health and Multimorbidity, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jacob Williams
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Haoyu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
5
|
Hou W, Liu Y, Hao X, Qi J, Jiang Y, Huang S, Zeng P. Relatively independent and complementary roles of family history and polygenic risk score in age at onset and incident cases of 12 common diseases. Soc Sci Med 2025; 371:117942. [PMID: 40073521 DOI: 10.1016/j.socscimed.2025.117942] [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: 10/11/2024] [Revised: 02/15/2025] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
Few studies have systematically compared the overlap and complementarity of family history (FH) and polygenic risk score (PRS) in terms of disease risk. We here investigated the impacts of FH and PRS on the risk of incident diseases or age at disease onset, as well as their clinical value in risk prediction. We analyzed 12 diseases in the prospective cohort study of UK Biobank (N = 461,220). First, restricted mean survival time analysis was performed to evaluate the influences of FH and PRS on age at onset. Then, Cox proportional hazards model was employed to estimate the effects of FH and PRS on the incident risk. Finally, prediction models were constructed to examine the clinical value of FH and PRS in the incident disease risk. Compared to negative FH, positive FH led to an earlier onset, with an average of 2.29 years earlier between the top and bottom 2.5% PRSs and high blood pressure showing the greatest difference of 6.01 years earlier. Both FH and PRS were related to higher incident risk; but they only exhibited weak interactions on high blood pressure and Alzheimer's disease/dementia, and provided relatively independent and partially complementary information on disease susceptibility, with PRS explaining 7.0% of the FH effect but FH accounting for only 1.1% of the PRS effect for incident cases. Further, FH and PRS showed additional predictive value in risk evaluation, with breast cancer showing the greatest improvement (31.3%). FH and PRS significantly affect a variety of diseases, and they are not interchangeable measures of genetic susceptibility, but instead offer largely independent and partially complementary information. Incorporating FH, PRS, and clinical risk factors simultaneously leads to the greatest predictive value for disease risk assessment.
Collapse
Affiliation(s)
- Wenyan Hou
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Yuxin Liu
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Jike Qi
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Yuchen Jiang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Shuiping Huang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China; Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China; Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
6
|
Zhang S, Wang T, Zeng P. Associations of Maternal Smoking During Pregnancy and Genetic Susceptibility with Incident Asthma from a Cohort Study. PREVENTION SCIENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR PREVENTION RESEARCH 2025; 26:343-354. [PMID: 40045075 DOI: 10.1007/s11121-025-01793-z] [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] [Accepted: 02/20/2025] [Indexed: 05/10/2025]
Abstract
Maternal smoking during pregnancy exhibited an adverse influence on asthma, but its joint effect with genetic factors remained elusive. Moreover, whether there existed a moderating role of genetic susceptibility in this effect on childhood/adolescent-onset asthma (COA) and adult-out asthma (AOA) was unknown. We employed the UK Biobank cohort to estimate the effect of maternal smoking during pregnancy on the risk of offspring asthma (41,828 AOA and 15,120 COA). We investigated genetic influence on asthma and assessed the moderating role of genetic susceptibility in this effect by incorporating polygenetic risk score (PRS) and performing a stratified analysis in distinct genetic risk populations. Hazard ratio (HR) and 95% confidence intervals (CIs) were reported. We found that participants whose mother smoked during pregnancy were more likely to occur asthma (HR = 1.14, 95%CIs 1.12 ~ 1.16), with similar effects for AOA and COA. Additionally, we observed a significant association between genetic factors and asthma (HR = 1.70, 95%CIs 1.66 ~ 1.74), with a higher genetic influence on COA (HR = 2.16, 95%CIs 2.09 ~ 2.23) compared to AOA (HR = 1.84, 95%CIs 1.76 ~ 1.93). Furthermore, we revealed that genetic factors could modify the effect of maternal smoking during pregnancy on asthma especially among childhood and adolescents, with participants having high genetic risk versus low genetic risk (HR = 1.13 vs. 1.02, P = 0.035). We provided supportive evidence that maternal smoking during pregnancy and the genetic factors increased the risk of offspring asthma in whole population. We further revealed that genetic susceptibility exerted more pronounced influence on COA compared to AOA, and played a moderating role in this effect.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ting Wang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
7
|
Yao Q, Gorevic PD, Gibson G. Genetically Transitional Disease and the Road to Personalized Medicine. Genes (Basel) 2025; 16:401. [PMID: 40282361 PMCID: PMC12026687 DOI: 10.3390/genes16040401] [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: 03/01/2025] [Revised: 03/22/2025] [Accepted: 03/28/2025] [Indexed: 04/29/2025] Open
Abstract
Genetically transitional disease (GTD) is emerging as a new concept in genomic medicine to straddle between the traditional binary classification of monogenic and polygenic disease. Genetic testing result reports in molecular laboratories have been predicated on the monogenic disease model, which focuses on pathogenic and likely pathogenic variants. While variants of uncertain significance (VUS) are reported by laboratories, there are challenges with regard to their clinical application so that these variants are often dismissed by ordering physicians. Unlike Mendelian disorders, where genetic variants are of high penetrance and highly probabilistic, the GTD concept is employed to highlight the impact of low-to-moderate effect gene variants whose influence on disease is modified by the genetic background. The GTD concept may explain health conditions associated with variants that are necessary but not sufficient for pathogenesis, lying in the mid gray zone between Mendelian and polygenic diseases. Although VUSs may not reach the level of pathogenicity based on American College of Medical Genetics and Genomics guidelines, they could be provisionally classified as GTD-associated variants to annotate and interpret the relationship between VUS and human genetic disease. The appropriate implementation of the GTD concept could impact patient care and research by focusing attention on the individual variability of responses in various diseases.
Collapse
Affiliation(s)
- Qingping Yao
- Division of Rheumatology, Allergy, and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA
| | - Peter D. Gorevic
- Division of Rheumatology, Allergy, and Immunology, Stony Brook University Renaissance School of Medicine, Stony Brook, NY 11794, USA
| | - Greg Gibson
- Center for Integrative Genomics, School of Biology, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| |
Collapse
|
8
|
Lorincz-Comi N, Cheng F. Bayesian estimation of shared polygenicity identifies drug targets and repurposable medicines for human complex diseases. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.17.25324106. [PMID: 40166559 PMCID: PMC11957083 DOI: 10.1101/2025.03.17.25324106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Complex diseases may share portions of their polygenic architectures which can be leveraged to identify drug targets with low off-target potential or repurposable candidates. However, the literature lacks methods which can make these inferences at scale using publicly available data. Methods We introduce a Bayesian model to estimate the polygenic structure of a trait using only gene-based association test statistics from GWAS summary data and returns gene-level posterior risk probabilities (PRPs). PRPs were used to infer shared polygenicity between 496 trait pairs and we introduce measures that can prioritize drug targets with low off-target effects or drug repurposing potential. Results Across 32 traits, we estimated that 69.5 to 97.5% of disease-associated genes are shared between multiple traits, and the estimated number of druggable genes that were only associated with a single disease ranged from 1 (multiple sclerosis) to 59 (schizophrenia). Estimating the shared genetic architecture of ALS with all other traits identified the KIT gene as a potentially harmful drug target because of its deleterious association with triglycerides, but also identified TBK1 and SCN11B as putatively safer because of their non-association with any of the other 31 traits. We additionally found 21 genes which are candidate repourposable targets for Alzheimer's disease (AD) (e.g., PLEKHA1, PPIB) and 5 for ALS (e.g., GAK, DGKQ). Conclusions The sets of candidate drug targets which have limited off-target potential are generally smaller compared to the sets of pleiotropic and putatively repurposable drug targets, but both represent promising directions for future experimental studies.
Collapse
Affiliation(s)
- Noah Lorincz-Comi
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Feixiong Cheng
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| |
Collapse
|
9
|
Plomin R, Vassos E. What clinicians should know about the contribution of modern behavioral genetics to psychiatric problems. Psychol Med 2025; 55:e83. [PMID: 40079100 PMCID: PMC12055018 DOI: 10.1017/s0033291725000273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/19/2025] [Accepted: 01/27/2025] [Indexed: 03/14/2025]
Affiliation(s)
- Robert Plomin
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| | - Evangelos Vassos
- Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, UK
| |
Collapse
|
10
|
Wang YG, Huang CC, Yeh TC, Chen WT, Chang WC, Singh AB, Yeh CB, Hung YJ, Hung KS, Chang HA. Novel ABCD1 and MTHFSD Variants in Taiwanese Bipolar Disorder: A Genetic Association Study. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:486. [PMID: 40142297 PMCID: PMC11943623 DOI: 10.3390/medicina61030486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025]
Abstract
Background and Objectives: In recent years, bipolar disorder (BD), a multifaceted mood disorder marked by severe episodic mood fluctuations, has been shown to have an impact on disability-adjusted life years (DALYs). The increasing prevalence of BD highlights the need for better diagnostic tools, particularly those involving genetic insights. Genetic association studies can play a crucial role in identifying variations linked to BD, shedding light on its genetic underpinnings and potential therapeutic targets. This study aimed to identify novel genetic variants associated with BD in the Taiwanese Han population and to elucidate their potential roles in disease pathogenesis. Materials and Methods: Genotyping was conducted using the Taiwan Precision Medicine Array (TPM Array) on 128 BD patients and 26,122 control subjects. Following quality control, 280,177 single nucleotide polymorphisms (SNPs) were analyzed via chi-square tests, and linkage disequilibrium (LD) analyses were employed to examine the associations among key SNPs. Results: Eleven SNPs reached significance (p < 10-5), with the variant rs11156606 in the ABCD1 gene-implicated in fatty acid metabolism-emerging as a prominent finding. LD analysis revealed that rs11156606 is strongly linked with rs73640819, located in the 3' untranslated region, suggesting a regulatory role in gene expression. Additionally, rs3829533 in the MTHFSD gene was found to be in strong LD with the missense variants rs3751800 and rs3751801, indicating potential alterations in protein function. Conclusion: These findings enhance the genetic understanding of BD within a Taiwanese cohort by identifying novel risk-associated variants and support the potential for using these markers in early diagnosis and targeted therapeutic strategies.
Collapse
Affiliation(s)
- Yi-Guang Wang
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-G.W.); (C.-C.H.); (T.-C.Y.); (W.-T.C.); (C.-B.Y.)
| | - Chih-Chung Huang
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-G.W.); (C.-C.H.); (T.-C.Y.); (W.-T.C.); (C.-B.Y.)
| | - Ta-Chuan Yeh
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-G.W.); (C.-C.H.); (T.-C.Y.); (W.-T.C.); (C.-B.Y.)
| | - Wan-Ting Chen
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-G.W.); (C.-C.H.); (T.-C.Y.); (W.-T.C.); (C.-B.Y.)
| | - Wei-Chou Chang
- Department of Radiology, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Ajeet B. Singh
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT), School of Medicine, Barwon Health, Deakin University, Geelong 3220, Australia;
| | - Chin-Bin Yeh
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-G.W.); (C.-C.H.); (T.-C.Y.); (W.-T.C.); (C.-B.Y.)
| | - Yi-Jen Hung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan;
| | - Kuo-Sheng Hung
- Center for Precision Medicine and Genomics, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan
| | - Hsin-An Chang
- Department of Psychiatry, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei 11490, Taiwan; (Y.-G.W.); (C.-C.H.); (T.-C.Y.); (W.-T.C.); (C.-B.Y.)
| |
Collapse
|
11
|
Hudson O, Brawner J. Using genome-wide associations and host-by-pathogen predictions to identify allelic interactions that control disease resistance. THE PLANT GENOME 2025; 18:e70006. [PMID: 39994874 PMCID: PMC11850958 DOI: 10.1002/tpg2.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 02/26/2025]
Abstract
Characterizing the molecular mechanisms underlying disease symptom expression has been used to improve human health and disease resistance in crops and animal breeds. Quantitative trait loci and genome-wide association studies (GWAS) are widely used to identify genomic regions that are involved in disease progression. This study extends traditional GWAS significance tests of host and pathogen marker main effects by utilizing dual-genome reaction norm models to evaluate the importance of host-single nucleotide polymorphism (SNP) by pathogen-SNP interactions. Disease symptom severity data from Fusarium ear rot (FER) on maize (Zea mays L.) is used to demonstrate the use of both genomes in genomic selection models for breeding and the identification of loci that interact across organisms to impact FER disease development. Dual genome prediction models improved heritability estimates, error variances, and model accuracy while providing predictions for host-by-pathogen interactions that may be used to test the significance of SNP-SNP interactions. Independent GWAS for maize and Fusarium populations identified significantly associated loci and predictions that were used to evaluate the importance of interactions using two different association tests. Predictions from dual genome models were used to evaluate the significance of the SNP-SNP interactions that may be associated with population structure or polygenic effects. As well, association tests incorporating host and pathogen markers in models that also included genomic relationship matrices were used to account for population structure. Subsequent evaluation of protein-protein interactions from candidate genes near the interacting SNPs provides a further in silico evaluation method to expedite the identification of interacting genes.
Collapse
Affiliation(s)
- Owen Hudson
- Department of Plant PathologyUniversity of FloridaGainesvilleFloridaUSA
| | - Jeremy Brawner
- Department of Plant PathologyUniversity of FloridaGainesvilleFloridaUSA
- Genics Ltd.Saint LuciaQueenslandAustralia
| |
Collapse
|
12
|
Doyle AE, Bearden CE, Gur RE, Ledbetter DH, Martin CL, McCoy TH, Pasaniuc B, Perlis RH, Smoller JW, Davis LK. Advancing Mental Health Research Through Strategic Integration of Transdiagnostic Dimensions and Genomics. Biol Psychiatry 2025; 97:450-460. [PMID: 39424167 DOI: 10.1016/j.biopsych.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Genome-wide studies are yielding a growing catalog of common and rare variants that confer risk for psychopathology. However, despite representing unprecedented progress, emerging data also indicate that the full promise of psychiatric genetics-including understanding pathophysiology and improving personalized care-will not be fully realized by targeting traditional dichotomous diagnostic categories. The current article provides reflections on themes that emerged from a 2021 National Institute of Mental Health-sponsored conference convened to address strategies for the evolving field of psychiatric genetics. As anticipated by the National Institute of Mental Health's Research Domain Criteria framework, multilevel investigations of dimensional and transdiagnostic phenotypes, particularly when integrated with biobanks and big data, will be critical to advancing knowledge. The path forward will also require more diverse representation in source studies. Additionally, progress will be catalyzed by a range of converging approaches, including capitalizing on computational methods, pursuing biological insights, working within a developmental framework, and engaging health care systems and patient communities.
Collapse
Affiliation(s)
- Alysa E Doyle
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| | - Carrie E Bearden
- Departments of Psychiatry and Biobehavioral Sciences & Psychology, University of California at Los Angeles, Los Angeles, California
| | - Raquel E Gur
- Departments of Psychiatry, Neurology and Radiology, Perelman School of Medicine, University of Pennsylvania, and the Lifespan Brain Institute of Children's Hospital of Philadelphia and Penn Medicine, Philadelphia, Pennsylvania
| | - David H Ledbetter
- Departments of Pediatrics and Psychiatry, University of Florida College of Medicine, Jacksonville, Florida
| | - Christa L Martin
- Geisinger Autism & Developmental Medicine Institute, Lewisburg, Pennsylvania
| | - Thomas H McCoy
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bogdan Pasaniuc
- Departments of Computational Medicine, Pathology and Laboratory Medicine, and Human Genetics, University of California at Los Angeles, Los Angeles, California
| | - Roy H Perlis
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Jordan W Smoller
- Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Lea K Davis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
13
|
Foden CJ, Durant K, Mellet J, Joubert F, van Rensburg J, Masemola K, Velaphi SC, Nakwa FL, Horn AR, Pillay S, Kali G, Coetzee M, Ballot DE, Kalua T, Babbo C, Pepper MS. Genetic Variants Associated with Suspected Neonatal Hypoxic Ischaemic Encephalopathy: A Study in a South African Context. Int J Mol Sci 2025; 26:2075. [PMID: 40076698 PMCID: PMC11900005 DOI: 10.3390/ijms26052075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Neonatal encephalopathy suspected to be due to hypoxic ischaemic encephalopathy (NESHIE) carries the risk of death or severe disability (cognitive defects and cerebral palsy). Previous genetic studies on NESHIE have predominantly focused on exomes or targeted genes. The objective of this study was to identify genetic variants associated with moderate-severe NESHIE through whole-genome, unbiased analysis. Variant filtering and prioritization were performed, followed by association testing both on a case-control basis and to compare the grades of severity and/or progression. Association testing on neonates with NESHIE (N = 172) and ancestry-matched controls (N = 288) produced 71 significant genetic variants (false discovery rate corrected p-value < 6.2 × 10-4), all located in non-coding regions and not previously implicated in NESHIE. Disease-associated variants in non-coding regions are considered to affect regulatory functions, possibly by modifying gene expression, promoters, enhancers, or DNA structure. The most significant variant was at position 6:162010973 in the Parkin RBR E3 ubiquitin protein ligase (PRKN) intron. Intronic variants were also identified in genes involved in inflammatory processes (SLCO3A1), DNA repair (ZGRF1), synaptogenesis (CNTN5), haematopoiesis (ASXL2), and the transcriptional response to hypoxia (PADI4). Ten variants were associated with a higher severity or lack of improvement in NESHIE, including one in ADAMTS3, which encodes a procollagen amino protease with a role in angiogenesis and lymphangiogenesis. This analysis represents one of the first efforts to analyze whole-genome data to investigate the genetic complexity of NESHIE in diverse ethnolinguistic groups of African origin and provides direction for further study.
Collapse
Affiliation(s)
- Caroline J. Foden
- Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa; (C.J.F.); (J.M.); (J.v.R.); (T.K.); (C.B.)
| | | | - Juanita Mellet
- Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa; (C.J.F.); (J.M.); (J.v.R.); (T.K.); (C.B.)
| | - Fourie Joubert
- Centre for Bioinformatics and Computational Biology, Genomics Research Institute, Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Pretoria 0002, South Africa;
| | - Jeanne van Rensburg
- Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa; (C.J.F.); (J.M.); (J.v.R.); (T.K.); (C.B.)
| | - Khomotso Masemola
- Department of Paediatrics and Child Health, Kalafong Hospital and Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa;
| | - Sithembiso C. Velaphi
- Department of Paediatrics and Child Health, Chris Hani Baragwanath Academic Hospital, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; (S.C.V.); (F.L.N.)
| | - Firdose L. Nakwa
- Department of Paediatrics and Child Health, Chris Hani Baragwanath Academic Hospital, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; (S.C.V.); (F.L.N.)
| | - Alan R. Horn
- Division of Neonatal Medicine, Department of Paediatrics and Child Health, Groote Schuur Hospital, University of Cape Town, Cape Town 7701, South Africa; (A.R.H.); (S.P.)
| | - Shakti Pillay
- Division of Neonatal Medicine, Department of Paediatrics and Child Health, Groote Schuur Hospital, University of Cape Town, Cape Town 7701, South Africa; (A.R.H.); (S.P.)
| | - Gugu Kali
- Tygerberg Hospital Neonatal Unit, Department of Paediatrics and Child Health, Stellenbosch University, Cape Town 7600, South Africa;
| | - Melantha Coetzee
- Division of Neonatology, Department of Paediatrics and Child Health, Steve Biko Academic Hospital, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa;
| | - Daynia E. Ballot
- Department of Paediatrics and Child Health, Charlotte Maxeke Johannesburg Academic Hospital, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa;
| | - Thumbiko Kalua
- Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa; (C.J.F.); (J.M.); (J.v.R.); (T.K.); (C.B.)
| | - Carina Babbo
- Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa; (C.J.F.); (J.M.); (J.v.R.); (T.K.); (C.B.)
| | - Michael S. Pepper
- Institute for Cellular and Molecular Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0084, South Africa; (C.J.F.); (J.M.); (J.v.R.); (T.K.); (C.B.)
| |
Collapse
|
14
|
Musial A, Foye U, Kakar S, Jewell T, Treasure J, Kalsi G, Smith I, Meldrum L, Bristow S, Marsh I, Malouf CM, Arora J, Davies H, Dutta R, Schmidt U, Breen G, Herle M. Genomic links between symptoms of eating disorders and suicidal ideation. Eur Psychiatry 2025; 68:1-31. [PMID: 39967258 PMCID: PMC11883781 DOI: 10.1192/j.eurpsy.2025.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/08/2025] [Accepted: 02/02/2025] [Indexed: 02/20/2025] Open
Abstract
Eating disorders, including anorexia nervosa, bulimia nervosa and binge eating disorder, are psychiatric conditions associated with high mortality rates, particularly due to suicide. Although eating disorders are strongly associated with suicidal ideation, attempts, and fatalities, the precise relationship between these conditions remains poorly understood. While substantial genetic influences have been identified for both eating disorders and suicidality, the shared genetics contributing to their co-occurrence remain unclear. In this study, we utilized a multivariate approach to examine the shared genetic architecture of eating disorder symptoms, suicidal thoughts and behaviors in ~20,000 participants from the COVID-19 Psychiatry and Neurological Genetics (COPING) study. We applied individual-level structural equation modeling to explore the factor structure underlying eating disorder symptoms and suicidal ideation, followed by genetic correlation analyses. We modeled the general factor of susceptibility to eating disorders and suicidal ideation that was as strongly genetically influenced as both conditions, with mean SNP heritability of 9%. Importantly, despite the frequent co-occurrence of eating disorders with other psychiatric conditions, our findings highlight the specificity of the relationship between eating disorders and suicidality, independent of other co-occurring psychopathology, such as depression and anxiety. This specificity highlights the need for targeted approaches in understanding the shared susceptibility factors.
Collapse
Affiliation(s)
- Agnieszka Musial
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Una Foye
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Saakshi Kakar
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Tom Jewell
- Department of Mental Health Nursing, Florence Nightingale Faculty of Nursing, Midwifery & Palliative Care, King’s College London, London, United Kingdom
| | - Janet Treasure
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Gursharan Kalsi
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Iona Smith
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Laura Meldrum
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Shannon Bristow
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Ian Marsh
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Chelsea Mika Malouf
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Jahnavi Arora
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Helena Davies
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Rina Dutta
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Ulrike Schmidt
- Department of Psychological Medicine, School of Academic Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | - Gerome Breen
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
- United Kingdom National Institute for Health and Care Research (NIHR) Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, United Kingdom
| | - Moritz Herle
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| |
Collapse
|
15
|
Li Y, Coelho A, Li Z, Alsved M, Li Q, Xu R, Luo H, Liang D, Xu J, Nandakumar KS, Meng L, Löndahl J, Holmdahl R. The systemic lupus erythematosus-associated NCF1 90H allele synergizes with viral infection to cause mouse lupus but also limits virus spread. Nat Commun 2025; 16:1593. [PMID: 39939342 PMCID: PMC11822037 DOI: 10.1038/s41467-025-56857-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
Studying how single nucleotide polymorphisms (SNPs) crosstalk with non-autologous factors to cause complex autoimmune diseases is challenging. An amino acid replacement in the neutrophil cytosolic factor 1 (NCF1-339/NCF1R90H) leading to lower reactive oxygen species induction has been reported as the major SNP for systemic lupus erythematosus (SLE). Here we show that infection with the murine norovirus (MNV) contributes to the induction of lupus in Ncf190H mice. Mutant NCF190H upregulates the IFN-α/JAK1/STAT1 pathway in macrophages and anti-MNV-antibody production. In parallel, the MNV infection of NCF190H mice upregulates Toll-like receptor 7 in macrophages, plasmacytoid dendritic cells and B220+ splenocytes, thereby promoting germinal center formation and lupus-associated autoantibodies production. These compounded effects lead to protection against MNV infection but also glomeruloneph ritis with proteinuria and lupus arthritis in the absence of chemical inducers such as pristane. Our data thus suggest that this SLE-associated SNP, NCF190H, synergizes with MNV infection to induce the development of mouse lupus.
Collapse
Grants
- The EU COSMIC Marie Curie grant (765158), the Swedish Research Council (2023-06482), Southern Medical University (SMU) grant (C1034211), the Natural Science Foundation of China (No.32070913, 82471830, W2431021), Vetenskapsrådet (VR) (2024-02575), NovoNordisk (NNF24OC0090035), Leo Foundation (LF-OC-22-001023), Cancer foundation (22 2350 Pj 01 H), and KAW (2019.0059).
- The KI Foundation for Virus Research (2023-00122), KI Foundation funds for rheumatology research (2023-02710)
- Science and Technology Major Project District-School Cooperation Outstanding Youth Fund (Shenzhen Nanshan District Health System (NSZD)(NSZD2023062)
Collapse
Affiliation(s)
- Yanpeng Li
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
- SMU-KI United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Ana Coelho
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Zhilei Li
- Clinical Pharmacy Division, Department of Pharmacy, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Malin Alsved
- Division of Ergonomics and Aerosol Technology, Faculty of Engineering, Lund University, Lund, Sweden
| | - Qixing Li
- SMU-KI United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Rui Xu
- SMU-KI United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Huqiao Luo
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Dongxia Liang
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Second Affiliated Hospital of Xi' an Jiaotong University (Xibei Hospital), Xi' an, China
| | - Jing Xu
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China
| | - Kutty Selva Nandakumar
- SMU-KI United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Liesu Meng
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Second Affiliated Hospital of Xi' an Jiaotong University (Xibei Hospital), Xi' an, China
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China
| | - Jakob Löndahl
- Division of Ergonomics and Aerosol Technology, Faculty of Engineering, Lund University, Lund, Sweden
| | - Rikard Holmdahl
- Medical Inflammation Research, Division of Immunology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.
- SMU-KI United Medical Inflammation Center, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Second Affiliated Hospital of Xi' an Jiaotong University (Xibei Hospital), Xi' an, China.
| |
Collapse
|
16
|
Liu X, Crawford L, Ramachandran S. ML-MAGES: A machine learning framework for multivariate genetic association analyses with genes and effect size shrinkage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637655. [PMID: 39990474 PMCID: PMC11844528 DOI: 10.1101/2025.02.11.637655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
A fundamental goal of genetics is to identify which and how genetic variants are associated with a trait, often using the regression results from genome-wide association (GWA) studies. Important methodological challenges are accounting for inflation in GWA effect estimates as well as investigating more than one trait simultaneously. We leverage machine learning approaches for these two challenges, developing a computationally efficient method called ML-MAGES. First, we shrink the inflation in GWA effect sizes caused by non-independence among variants using neural networks. We then cluster variant associations among multiple traits via variational inference. We compare the performance of shrinkage via neural networks to regularized regression and fine-mapping, two approaches used for addressing inflated effects but dealing with variants in focal regions of different sizes. Our neural network shrinkage outperforms both methods in approximating the true effect sizes in simulated data. Our infinite mixture clustering approach offers a flexible, data-driven way to distinguish different types of associations-trait-specific, shared across traits, or spurious-among multiple traits based on their regularized effects. Clustering applied to our neural network shrinkage results also produces consistently higher precision and recall for distinguishing gene-level associations in simulations. We demonstrate the application of ML-MAGES on association analyses of two quantitative traits and two binary traits in the UK Biobank (genetic and phenotypic data from 500,000 residents of the UK). Our identified associated genes from single-trait enrichment tests overlap with those having known relevant biological processes to the traits. Besides trait-specific associations, ML-MAGES identifies several variants with shared multi-trait associations, suggesting putative shared genetic architecture.
Collapse
Affiliation(s)
- Xiran Liu
- Brown University, Providence, RI 02906, USA
| | - Lorin Crawford
- Brown University, Providence, RI 02906, USA
- Microsoft Research, Cambridge, MA 02142, USA
| | | |
Collapse
|
17
|
Winters JLG, Piekos JA, Hellwege JN, Dikilitas O, Kullo IJ, Schaid DJ, Edwards TL, Velez Edwards DR. Constructing a multi-ancestry polygenic risk score for uterine fibroids using publicly available data highlights need for inclusive genetic research. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2025; 30:268-280. [PMID: 39670376 PMCID: PMC11731894 DOI: 10.1142/9789819807024_0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Uterine leiomyomata, or fibroids, are common gynecological tumors causing pelvic and menstrual symptoms that can negatively affect quality of life and child-bearing desires. As fibroids grow, symptoms can intensify and lead to invasive treatments that are less likely to preserve fertility. Identifying individuals at highest risk for fibroids can aid in access to earlier diagnoses. Polygenic risk scores (PRS) quantify genetic risk to identify those at highest risk for disease. Utilizing the PRS software PRS-CSx and publicly available genome-wide association study (GWAS) summary statistics from FinnGen and Biobank Japan, we constructed a multi-ancestry (META) PRS for fibroids. We validated the META PRS in two cross-ancestry cohorts. In the cross-ancestry Electronic Medical Record and Genomics (eMERGE) Network cohort, the META PRS was significantly associated with fibroid status and exhibited 1.11 greater odds for fibroids per standard deviation increase in PRS (95% confidence interval [CI]: 1.05 - 1.17, p = 5.21x10-5). The META PRS was validated in two BioVU cohorts: one using ICD9/ICD10 codes and one requiring imaging confirmation of fibroid status. In the ICD cohort, a standard deviation increase in the META PRS increased the odds of fibroids by 1.23 (95% CI: 1.15 - 1.32, p = 9.68x10-9), while in the imaging cohort, the odds increased by 1.26 (95% CI: 1.18 - 1.35, p = 2.40x10-11). We subsequently constructed single ancestry PRS for FinnGen (European ancestry [EUR]) and Biobank Japan (East Asian ancestry [EAS]) using PRS-CS and discovered a nominally significant association in the eMERGE cohort within fibroids and EAS PRS but not EUR PRS (95% CI: 1.09 - 1.20, p = 1.64x10-7). These findings highlight the strong predictive power of multi-ancestry PRS over single ancestry PRS. This study underscores the necessity of diverse population inclusion in genetic research to ensure precision medicine benefits all individuals equitably.
Collapse
Affiliation(s)
- Jessica L G Winters
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Jacqueline A Piekos
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Jacklyn N Hellwege
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37203, USA
| | - Ozan Dikilitas
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Iftikhar J Kullo
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel J Schaid
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Todd L Edwards
- Division of Epidemiology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37203, USA,
| | - Digna R Velez Edwards
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37203, USA,
| |
Collapse
|
18
|
Crouch DJM, Inshaw JRJ, Robertson CC, Ng E, Zhang J, Chen W, Onengut‐Gumuscu S, Cutler AJ, Sidore C, Cucca F, Pociot F, Concannon P, Rich SS, Todd JA. Bayesian Effect Size Ranking to Prioritise Genetic Risk Variants in Common Diseases for Follow-Up Studies. Genet Epidemiol 2025; 49:e22608. [PMID: 39749473 PMCID: PMC11696485 DOI: 10.1002/gepi.22608] [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: 03/28/2024] [Revised: 09/10/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
Biological datasets often consist of thousands or millions of variables, e.g. genetic variants or biomarkers, and when sample sizes are large it is common to find many associated with an outcome of interest, for example, disease risk in a GWAS, at high levels of statistical significance, but with very small effects. The False Discovery Rate (FDR) is used to identify effects of interest based on ranking variables according to their statistical significance. Here, we develop a complementary measure to the FDR, the priorityFDR, that ranks variables by a combination of effect size and significance, allowing further prioritisation among a set of variables that pass a significance or FDR threshold. Applying to the largest GWAS of type 1 diabetes to date (15,573 cases and 158,408 controls), we identified 26 independent genetic associations, including two newly-reported loci, with qualitatively lower priorityFDRs than the remaining 175 signals. We detected putatively causal type 1 diabetes risk genes using Mendelian Randomisation, and found that these were located disproportionately close to low priorityFDR signals (p = 0.005), as were genes in the IL-2 pathway (p = 0.003). Selecting variables on both effect size and significance can lead to improved prioritisation for mechanistic follow-up studies from genetic and other large biological datasets.
Collapse
Grants
- DP3 DK111906 NIDDK NIH HHS
- T32 LM012416 NLM NIH HHS
- U01 DK062418 NIDDK NIH HHS
- Wellcome Trust
- This work was funded by JDRF grants 9-2011-253, 5-SRA-2015-130-A-N and 4-SRA-2017-473-A-N, and Wellcome grants 091157/Z/10/Z and 107212/Z/15/Z, to the Diabetes and Inflammation Laboratory, University of Oxford, Fondazione di Sardegna grant U1301.2015/AI.1157.BE to Francesco Cucca, NIDDK grants U01 DK062418 and DP3 DK111906 and US National Library of Medicine grant T32 LM012416.
- This work was funded by JDRF grants 9‐2011‐253, 5‐SRA‐2015‐130‐A‐N and 4‐SRA‐2017‐473‐A‐N, and Wellcome grants 091157/Z/10/Z and 107212/Z/15/Z, to the Diabetes and Inflammation Laboratory, University of Oxford, Fondazione di Sardegna grant U1301.2015/AI.1157.BE to Francesco Cucca, NIDDK grants U01 DK062418 and DP3 DK111906 and US National Library of Medicine grant T32 LM012416.
Collapse
Affiliation(s)
- Daniel J. M. Crouch
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Jamie R. J. Inshaw
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | | | - Esther Ng
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
- Nuffield Department of OrthopaedicsKennedy Institute of Rheumatology, Rheumatology and Musculoskeletal SciencesUniversity of OxfordOxfordUK
| | - Jia‐Yuan Zhang
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Wei‐Min Chen
- Center for Public Health GenomicsUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Public Health SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Suna Onengut‐Gumuscu
- Center for Public Health GenomicsUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Public Health SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Antony J. Cutler
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| | - Carlo Sidore
- Institute for Research in Genetics and Biomedicine (IRGB)SardiniaItaly
| | - Francesco Cucca
- Institute for Research in Genetics and Biomedicine (IRGB)SardiniaItaly
| | - Flemming Pociot
- Department of PediatricsHerlev University HospitalCopenhagenDenmark
- Institute of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Clinical ResearchSteno Diabetes Center Copenhagen, Type 1 Diabetes BiologyGentofteDenmark
| | - Patrick Concannon
- Department of Pathology, Immunology, and Laboratory MedicineUniversity of FloridaGainesvilleFloridaUSA
- Genetics InstituteUniversity of FloridaGainesvilleFloridaUSA
| | - Stephen S. Rich
- Center for Public Health GenomicsUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Public Health SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - John A. Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Nuffield Department of MedicineCentre for Human Genetics, NIHR Oxford Biomedical Research CentreUniversity of OxfordOxfordUK
| |
Collapse
|
19
|
Cocoș R, Popescu BO. Scrutinizing neurodegenerative diseases: decoding the complex genetic architectures through a multi-omics lens. Hum Genomics 2024; 18:141. [PMID: 39736681 DOI: 10.1186/s40246-024-00704-7] [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: 10/05/2024] [Accepted: 12/10/2024] [Indexed: 01/01/2025] Open
Abstract
Neurodegenerative diseases present complex genetic architectures, reflecting a continuum from monogenic to oligogenic and polygenic models. Recent advances in multi-omics data, coupled with systems genetics, have significantly refined our understanding of how these data impact neurodegenerative disease mechanisms. To contextualize these genetic discoveries, we provide a comprehensive critical overview of genetic architecture concepts, from Mendelian inheritance to the latest insights from oligogenic and omnigenic models. We explore the roles of common and rare genetic variants, gene-gene and gene-environment interactions, and epigenetic influences in shaping disease phenotypes. Additionally, we emphasize the importance of multi-omics layers including genomic, transcriptomic, proteomic, epigenetic, and metabolomic data in elucidating the molecular mechanisms underlying neurodegeneration. Special attention is given to missing heritability and the contribution of rare variants, particularly in the context of pleiotropy and network pleiotropy. We examine the application of single-cell omics technologies, transcriptome-wide association studies, and epigenome-wide association studies as key approaches for dissecting disease mechanisms at tissue- and cell-type levels. Our review introduces the OmicPeak Disease Trajectory Model, a conceptual framework for understanding the genetic architecture of neurodegenerative disease progression, which integrates multi-omics data across biological layers and time points. This review highlights the critical importance of adopting a systems genetics approach to unravel the complex genetic architecture of neurodegenerative diseases. Finally, this emerging holistic understanding of multi-omics data and the exploration of the intricate genetic landscape aim to provide a foundation for establishing more refined genetic architectures of these diseases, enhancing diagnostic precision, predicting disease progression, elucidating pathogenic mechanisms, and refining therapeutic strategies for neurodegenerative conditions.
Collapse
Affiliation(s)
- Relu Cocoș
- Department of Medical Genetics, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania.
- Genomics Research and Development Institute, Bucharest, Romania.
| | - Bogdan Ovidiu Popescu
- Department of Clinical Neurosciences, 'Carol Davila' University of Medicine and Pharmacy, Bucharest, Romania.
| |
Collapse
|
20
|
Leve LD, Kanamori M, Humphreys KL, Jaffee SR, Nusslock R, Oro V, Hyde LW. The Promise and Challenges of Integrating Biological and Prevention Sciences: A Community-Engaged Model for the Next Generation of Translational Research. PREVENTION SCIENCE : THE OFFICIAL JOURNAL OF THE SOCIETY FOR PREVENTION RESEARCH 2024; 25:1177-1199. [PMID: 39225944 PMCID: PMC11652675 DOI: 10.1007/s11121-024-01720-8] [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] [Accepted: 08/11/2024] [Indexed: 09/04/2024]
Abstract
Beginning with the successful sequencing of the human genome two decades ago, the possibility of developing personalized health interventions based on one's biology has captured the imagination of researchers, medical providers, and individuals seeking health care services. However, the application of a personalized medicine approach to emotional and behavioral health has lagged behind the development of personalized approaches for physical health conditions. There is potential value in developing improved methods for integrating biological science with prevention science to identify risk and protective mechanisms that have biological underpinnings, and then applying that knowledge to inform prevention and intervention services for emotional and behavioral health. This report represents the work of a task force appointed by the Board of the Society for Prevention Research to explore challenges and recommendations for the integration of biological and prevention sciences. We present the state of the science and barriers to progress in integrating the two approaches, followed by recommended strategies that would promote the responsible integration of biological and prevention sciences. Recommendations are grounded in Community-Based Participatory Research approaches, with the goal of centering equity in future research aimed at integrating the two disciplines to ultimately improve the well-being of those who have disproportionately experienced or are at risk for experiencing emotional and behavioral problems.
Collapse
Affiliation(s)
- Leslie D Leve
- Prevention Science Institute, University of Oregon, Eugene, USA.
- Department of Counseling Psychology and Human Services, University of Oregon, Eugene, USA.
- Cambridge Public Health, University of Cambridge, Cambridge, UK.
| | - Mariano Kanamori
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, USA
| | - Kathryn L Humphreys
- Department of Psychology and Human Development, Vanderbilt University, Nashville, USA
| | - Sara R Jaffee
- Department of Psychology, University of Pennsylvania, Philadelphia, USA
| | - Robin Nusslock
- Department of Psychology & Institute for Policy Research, Northwestern University, Evanston, USA
| | - Veronica Oro
- Prevention Science Institute, University of Oregon, Eugene, USA
| | - Luke W Hyde
- Department of Psychology & Survey Research Center at the Institute for Social Research, University of Michigan, Ann Arbor, USA
| |
Collapse
|
21
|
Guzmán-Jiménez A, González-Muñoz S, Cerván-Martín M, Garrido N, Castilla JA, Gonzalvo MC, Clavero A, Molina M, Luján S, Santos-Ribeiro S, Vilches MÁ, Espuch A, Maldonado V, Galiano-Gutiérrez N, Santamaría-López E, González-Ravina C, Quintana-Ferraz F, Gómez S, Amorós D, Martínez-Granados L, Ortega-González Y, Burgos M, Pereira-Caetano I, Bulbul O, Castellano S, Romano M, Albani E, Bassas L, Seixas S, Gonçalves J, Lopes AM, Larriba S, Palomino-Morales RJ, Carmona FD, Bossini-Castillo L. A comprehensive study of common and rare genetic variants in spermatogenesis-related loci identifies new risk factors for idiopathic severe spermatogenic failure. Hum Reprod Open 2024; 2024:hoae069. [PMID: 39678461 PMCID: PMC11645127 DOI: 10.1093/hropen/hoae069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/11/2024] [Indexed: 12/17/2024] Open
Abstract
STUDY QUESTION Can genome-wide genotyping data be analysed using a hypothesis-driven approach to enhance the understanding of the genetic basis of severe spermatogenic failure (SPGF) in male infertility? SUMMARY ANSWER Our findings revealed a significant association between SPGF and the SHOC1 gene and identified three novel genes (PCSK4, AP3B1, and DLK1) along with 32 potentially pathogenic rare variants in 30 genes that contribute to this condition. WHAT IS KNOWN ALREADY SPGF is a major cause of male infertility, often with an unknown aetiology. SPGF can be due to either multifactorial causes, including both common genetic variants in multiple genes and environmental factors, or highly damaging rare variants. Next-generation sequencing methods are useful for identifying rare mutations that explain monogenic forms of SPGF. Genome-wide association studies (GWASs) have become essential approaches for deciphering the intricate genetic landscape of complex diseases, offering a cost-effective and rapid means to genotype millions of genetic variants. Novel methods have demonstrated that GWAS datasets can be used to infer rare coding variants that are causal for male infertility phenotypes. However, this approach has not been previously applied to characterize the genetic component of a whole case-control cohort. STUDY DESIGN SIZE DURATION We employed a hypothesis-driven approach focusing on all genetic variation identified, using a GWAS platform and subsequent genotype imputation, encompassing over 20 million polymorphisms and a total of 1571 SPGF patients and 2431 controls. Both common (minor allele frequency, MAF > 0.01) and rare (MAF < 0.01) variants were investigated within a total of 1797 loci with a reported role in spermatogenesis. This gene panel was meticulously assembled through comprehensive searches in the literature and various databases focused on male infertility genetics. PARTICIPANTS/MATERIALS SETTING METHODS This study involved a European cohort using previously and newly generated data. Our analysis consisted of three independent methods: (i) variant-wise association analyses using logistic regression models, (ii) gene-wise association analyses using combined multivariate and collapsing burden tests, and (iii) identification and characterisation of highly damaging rare coding variants showing homozygosity only in SPGF patients. MAIN RESULTS AND THE ROLE OF CHANCE The variant-wise analyses revealed an association between SPGF and SHOC1-rs12347237 (P = 4.15E-06, odds ratio = 2.66), which was likely explained by an altered binding affinity of key transcription factors in regulatory regions and the disruptive effect of coding variants within the gene. Three additional genes (PCSK4, AP3B1, and DLK1) were identified as novel relevant players in human male infertility using the gene-wise burden test approach (P < 5.56E-04). Furthermore, we linked a total of 32 potentially pathogenic and recessive coding variants of the selected genes to 35 different cases. LARGE SCALE DATA Publicly available via GWAS catalog (accession number: GCST90239721). LIMITATIONS REASONS FOR CAUTION The analysis of low-frequency variants presents challenges in achieving sufficient statistical power to detect genetic associations. Consequently, independent studies with larger sample sizes are essential to replicate our results. Additionally, the specific roles of the identified variants in the pathogenic mechanisms of SPGF should be assessed through functional experiments. WIDER IMPLICATIONS OF THE FINDINGS Our findings highlight the benefit of using GWAS genotyping to screen for both common and rare variants potentially implicated in idiopathic cases of SPGF, whether due to complex or monogenic causes. The discovery of novel genetic risk factors for SPGF and the elucidation of the underlying genetic causes provide new perspectives for personalized medicine and reproductive counselling. STUDY FUNDING/COMPETING INTERESTS This work was supported by the Spanish Ministry of Science and Innovation through the Spanish National Plan for Scientific and Technical Research and Innovation (PID2020-120157RB-I00) and the Andalusian Government through the research projects of 'Plan Andaluz de Investigación, Desarrollo e Innovación (PAIDI 2020)' (ref. PY20_00212) and 'Proyectos de Investigación aplicada FEDER-UGR 2023' (ref. C-CTS-273-UGR23). S.G.-M. was funded by the previously mentioned projects (ref. PY20_00212 and PID2020-120157RB-I00). A.G.-J. was funded by MCIN/AEI/10.13039/501100011033 and FSE 'El FSE invierte en tu futuro' (grant ref. FPU20/02926). IPATIMUP integrates the i3S Research Unit, which is partially supported by the Portuguese Foundation for Science and Technology (FCT), financed by the European Social Funds (COMPETE-FEDER) and National Funds (projects PEstC/SAU/LA0003/2013 and POCI-01-0145-FEDER-007274). S.S. is supported by FCT funds (10.54499/DL57/2016/CP1363/CT0019), ToxOmics-Centre for Toxicogenomics and Human Health, Genetics, Oncology and Human Toxicology, and is also partially supported by the Portuguese Foundation for Science and Technology (UIDP/00009/2020 and UIDB/00009/2020). S. Larriba received support from Instituto de Salud Carlos III (grant: DTS18/00101), co-funded by FEDER funds/European Regional Development Fund (ERDF)-a way to build Europe) and from 'Generalitat de Catalunya' (grant 2021SGR052). S. Larriba is also sponsored by the 'Researchers Consolidation Program' from the SNS-Dpt. Salut Generalitat de Catalunya (Exp. CES09/020). All authors declare no conflict of interest related to this study.
Collapse
Affiliation(s)
- Andrea Guzmán-Jiménez
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
| | - Sara González-Muñoz
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
| | - Miriam Cerván-Martín
- Institute of Parasitology and Biomedicine López-Neyra (IPBLN), CSIC, Granada, Spain
| | - Nicolás Garrido
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - José A Castilla
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Departamento de Anatomía y Embriología Humana, Facultad de Medicina, Universidad de Granada, Granada, Spain
| | - M Carmen Gonzalvo
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, Granada, Spain
| | - Ana Clavero
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, Granada, Spain
| | - Marta Molina
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Unidad de Reproducción, UGC Obstetricia y Ginecología, HU Virgen de las Nieves, Granada, Spain
| | - Saturnino Luján
- Servicio de Urología, Hospital Universitari i Politecnic La Fe e Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Samuel Santos-Ribeiro
- IVI-RMA Lisbon, Lisbon, Portugal
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - Miguel Ángel Vilches
- Ovoclinic & Ovobank, Clínicas de Reproducción Asistida y Banco de óvulos, Marbella, Málaga, Spain
| | - Andrea Espuch
- Hospital Universitario Torrecárdenas, Unidad de Reproducción Humana Asistida, Almería, Spain
| | - Vicente Maldonado
- UGC de Obstetricia y Ginecología, Complejo Hospitalario de Jaén, Jaén, Spain
| | | | | | - Cristina González-Ravina
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Fernando Quintana-Ferraz
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Susana Gómez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - David Amorós
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | | | | | - Miguel Burgos
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
| | - Iris Pereira-Caetano
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr Ricardo Jorge, Lisbon, Portugal
| | - Ozgur Bulbul
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Stefano Castellano
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Massimo Romano
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, Humanitas Research Hospital, IRCCS, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Elena Albani
- Division of Gynecology and Reproductive Medicine, Department of Gynecology, Fertility Center, Humanitas Research Hospital, IRCCS, Milan, Italy
| | - Lluís Bassas
- Laboratory of Seminology and Embryology, Andrology Service-Fundació Puigvert, Barcelona, Spain
| | - Susana Seixas
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
| | - João Gonçalves
- Departamento de Genética Humana, Instituto Nacional de Saúde Dr Ricardo Jorge, Lisbon, Portugal
- ToxOmics—Centro de Toxicogenómica e Saúde Humana, Nova Medical School, Lisbon, Portugal
| | - Alexandra M Lopes
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Porto, Portugal
- CGPP-IBMC—Centro de Genética Preditiva e Preventiva, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Sara Larriba
- Human Molecular Genetics Group, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
| | - Rogelio J Palomino-Morales
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
- Departamento de Bioquímica y Biología Molecular I, Universidad de Granada, Granada, Spain
| | - F David Carmona
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
| | - Lara Bossini-Castillo
- Departamento de Genética e Instituto de Biotecnología, Centro de Investigación Biomédica (CIBM), Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs. GRANADA, Granada, Spain
| |
Collapse
|
22
|
Hou W, Guan F, Chen W, Qi J, Huang S, Zeng P. Breastfeeding, genetic susceptibility, and the risk of asthma and allergic diseases in children and adolescents: a retrospective national population-based cohort study. BMC Public Health 2024; 24:3056. [PMID: 39501212 PMCID: PMC11539314 DOI: 10.1186/s12889-024-20501-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/23/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Asthma and allergic diseases (such as allergic rhinitis) are multifactorial chronic respiratory diseases, and have many common pathogenic mechanisms. This study aimed to assess the joint effects of breastfeeding and genetic susceptibility on asthma, allergic disease in children and adolescents and sought to examine whether the effect of breastfeeding was consistent under distinct levels of genetic risk. METHODS A total of 351,931 UK Biobank participants were analyzed. Firstly, Cox proportional hazards model was used to evaluate the relation between breastfeeding and asthma, allergic disease and their comorbidity. Next, we incorporated the polygenic risk score as an additional covariate into the model. Then, we explored the role of breastfeeding at each stage of asthma and allergic disease through a multi-state model. Meanwhile, several sensitivity analyses were conducted to evaluate the robustness of our results. Finally, we calculated the attributable protection and population attributable protection of breastfeeding. RESULTS Breastfeeding was related to a reduced risk of occurring asthma (adjusted hazard ratio [HR] = 0.89, 95% confidence interval [CI] 0.86 ~ 0.93), allergic disease (HR = 0.89, 95%CI 0.87 ~ 0.91) and comorbidity (HR = 0.89, 95%CI 0.83 ~ 0.94). The effect of breastfeeding was almost unchanged after considering PRS and did not substantially differ across distinct genetic risk levels. Breastfeeding showed a stronger risk-decreased impact on individuals who developed from allergic rhinitis to comorbidity (HR = 0.83, 95%CI 0.73 ~ 0.93). Further, the influence of breastfeeding was robust against covariates considered and the confounding influence of adolescent smoking. Finally, due to breastfeeding, 12.0%, 13.0% or 13.0% of the exposed population would not suffer from asthma, allergic diseases and the comorbidity, while 7.1%, 7.6% or 7.6% of the general population would not suffer from these diseases. CONCLUSIONS This study provided supportive evidence for the risk-reduced effect of breastfeeding on asthma, allergic diseases, and the comorbidity in children and adolescents, and further revealed that such an influence was consistent across distinct genetic risk levels.
Collapse
Affiliation(s)
- Wenyan Hou
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Fengjun Guan
- Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Wenying Chen
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jike Qi
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Shuiping Huang
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Ping Zeng
- Department of Biostatistics, School of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
- Jiangsu Engineering Research Center of Biological Data Mining and Healthcare Transformation, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
23
|
Ndong Sima CAA, Step K, Swart Y, Schurz H, Uren C, Möller M. Methodologies underpinning polygenic risk scores estimation: a comprehensive overview. Hum Genet 2024; 143:1265-1280. [PMID: 39425790 PMCID: PMC11522080 DOI: 10.1007/s00439-024-02710-0] [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] [Received: 08/13/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
Polygenic risk scores (PRS) have emerged as a promising tool for predicting disease risk and treatment outcomes using genomic data. Thousands of genome-wide association studies (GWAS), primarily involving populations of European ancestry, have supported the development of PRS models. However, these models have not been adequately evaluated in non-European populations, raising concerns about their clinical validity and predictive power across diverse groups. Addressing this issue requires developing novel risk prediction frameworks that leverage genetic characteristics across diverse populations, considering host-microbiome interactions and a broad range of health measures. One of the key aspects in evaluating PRS is understanding the strengths and limitations of various methods for constructing them. In this review, we analyze strengths and limitations of different methods for constructing PRS, including traditional weighted approaches and new methods such as Bayesian and Frequentist penalized regression approaches. Finally, we summarize recent advances in PRS calculation methods development, and highlight key areas for future research, including development of models robust across diverse populations by underlining the complex interplay between genetic variants across diverse ancestral backgrounds in disease risk as well as treatment response prediction. PRS hold great promise for improving disease risk prediction and personalized medicine; therefore, their implementation must be guided by careful consideration of their limitations, biases, and ethical implications to ensure that they are used in a fair, equitable, and responsible manner.
Collapse
Affiliation(s)
- Carene Anne Alene Ndong Sima
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Kathryn Step
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Yolandi Swart
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Haiko Schurz
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Caitlin Uren
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
- Centre for Bioinformatics and Computational Biology, Stellenbosch University, Cape Town, South Africa
| | - Marlo Möller
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa.
- Centre for Bioinformatics and Computational Biology, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
24
|
Curtis M, Colodro-Conde L, Medland SE, Gordon S, Martin NG, Wade TD, Cohen-Woods S. Anorexia nervosa polygenic risk, beyond diagnoses: relationship with adolescent disordered eating and behaviors in an Australian female twin population. Psychol Med 2024; 54:1-9. [PMID: 39439302 PMCID: PMC11536114 DOI: 10.1017/s0033291724001727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/05/2024] [Accepted: 06/21/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND It is well established that there is a substantial genetic component to eating disorders (EDs). Polygenic risk scores (PRSs) can be used to quantify cumulative genetic risk for a trait at an individual level. Recent studies suggest PRSs for anorexia nervosa (AN) may also predict risk for other disordered eating behaviors, but no study has examined if PRS for AN can predict disordered eating as a global continuous measure. This study aimed to investigate whether PRS for AN predicted overall levels of disordered eating, or specific lifetime disordered eating behaviors, in an Australian adolescent female population. METHODS PRSs were calculated based on summary statistics from the largest Psychiatric Genomics Consortium AN genome-wide association study to date. Analyses were performed using genome-wide complex trait analysis to test the associations between AN PRS and disordered eating global scores, avoidance of eating, objective bulimic episodes, self-induced vomiting, and driven exercise in a sample of Australian adolescent female twins recruited from the Australian Twin Registry (N = 383). RESULTS After applying the false-discovery rate correction, the AN PRS was significantly associated with all disordered eating outcomes. CONCLUSIONS Findings suggest shared genetic etiology across disordered eating presentations and provide insight into the utility of AN PRS for predicting disordered eating behaviors in the general population. In the future, PRSs for EDs may have clinical utility in early disordered eating risk identification, prevention, and intervention.
Collapse
Affiliation(s)
- Madeleine Curtis
- Discipline of Psychology, College of Education, Psychology, and Social Work, Flinders University, Adelaide, SA, Australia
- Blackbird Initiative, Flinders Institute for Mental Health and Wellbeing, Flinders University, Adelaide, SA, Australia
| | | | | | - Scott Gordon
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | | | - Tracey D. Wade
- Discipline of Psychology, College of Education, Psychology, and Social Work, Flinders University, Adelaide, SA, Australia
- Blackbird Initiative, Flinders Institute for Mental Health and Wellbeing, Flinders University, Adelaide, SA, Australia
| | - Sarah Cohen-Woods
- Discipline of Psychology, College of Education, Psychology, and Social Work, Flinders University, Adelaide, SA, Australia
- Blackbird Initiative, Flinders Institute for Mental Health and Wellbeing, Flinders University, Adelaide, SA, Australia
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
25
|
Ueda M. A brief clinical genetics review: stepwise diagnostic processes of a monogenic disorder-hypertriglyceridemia. Transl Pediatr 2024; 13:1828-1848. [PMID: 39524398 PMCID: PMC11543124 DOI: 10.21037/tp-24-131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/24/2024] [Indexed: 11/16/2024] Open
Abstract
The completion of the Human Genome Project and tremendous advances in automated high-throughput genetic analysis technologies have enabled explosive progress in the field of genetics, which resulted in countless discoveries of novel genes and pathways. Many phenotype- or disease-associated single nucleotide polymorphisms (SNPs) with a high statistical significance have been identified through numerous genome-wide association studies (GWAS), and various polygenic risk scoring (PRS) schemes have been proposed to identify individuals with a high risk for a certain trait or disorder. Meanwhile, medical education in genetics has lagged far behind, leaving many physicians and healthcare providers unprepared in the genomic era. Thus, there is an urgent need to educate physicians and healthcare providers with basic knowledge and skills in genetics. To facilitate this, some basic terminologies and concepts are discussed in this review. In addition, some important considerations in delineating and incorporating clinical genetic testing in the diagnosis and management of a monogenic disorder are illustrated in a stepwise fashion. Furthermore, the effects of disease-associated SNPs represented by a PRS scheme clearly demonstrated that even the phenotypes of a monogenic disorder due to the same pathogenic variant in family members are modulated by the polygenic background. In human genetics, despite these explosive advancements, we are still far from clearly deciphering the interplay of gene variants to effect unique characteristics in an individual. In addition, sophisticated genome or gene directed therapies are being investigated for numerous disorders. Therefore, evolution in the field of genetics is likely to continue into the foreseeable future. In the meantime, much emphasis should be placed on educating physicians and healthcare professionals to be well-versed and skillful in the clinical use of genetics so that they can fully embrace the new era of precision medicine.
Collapse
Affiliation(s)
- Masako Ueda
- Department of Medicine, The University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
26
|
Capalbo A, de Wert G, Mertes H, Klausner L, Coonen E, Spinella F, Van de Velde H, Viville S, Sermon K, Vermeulen N, Lencz T, Carmi S. Screening embryos for polygenic disease risk: a review of epidemiological, clinical, and ethical considerations. Hum Reprod Update 2024; 30:529-557. [PMID: 38805697 PMCID: PMC11369226 DOI: 10.1093/humupd/dmae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/25/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND The genetic composition of embryos generated by in vitro fertilization (IVF) can be examined with preimplantation genetic testing (PGT). Until recently, PGT was limited to detecting single-gene, high-risk pathogenic variants, large structural variants, and aneuploidy. Recent advances have made genome-wide genotyping of IVF embryos feasible and affordable, raising the possibility of screening embryos for their risk of polygenic diseases such as breast cancer, hypertension, diabetes, or schizophrenia. Despite a heated debate around this new technology, called polygenic embryo screening (PES; also PGT-P), it is already available to IVF patients in some countries. Several articles have studied epidemiological, clinical, and ethical perspectives on PES; however, a comprehensive, principled review of this emerging field is missing. OBJECTIVE AND RATIONALE This review has four main goals. First, given the interdisciplinary nature of PES studies, we aim to provide a self-contained educational background about PES to reproductive specialists interested in the subject. Second, we provide a comprehensive and critical review of arguments for and against the introduction of PES, crystallizing and prioritizing the key issues. We also cover the attitudes of IVF patients, clinicians, and the public towards PES. Third, we distinguish between possible future groups of PES patients, highlighting the benefits and harms pertaining to each group. Finally, our review, which is supported by ESHRE, is intended to aid healthcare professionals and policymakers in decision-making regarding whether to introduce PES in the clinic, and if so, how, and to whom. SEARCH METHODS We searched for PubMed-indexed articles published between 1/1/2003 and 1/3/2024 using the terms 'polygenic embryo screening', 'polygenic preimplantation', and 'PGT-P'. We limited the review to primary research papers in English whose main focus was PES for medical conditions. We also included papers that did not appear in the search but were deemed relevant. OUTCOMES The main theoretical benefit of PES is a reduction in lifetime polygenic disease risk for children born after screening. The magnitude of the risk reduction has been predicted based on statistical modelling, simulations, and sibling pair analyses. Results based on all methods suggest that under the best-case scenario, large relative risk reductions are possible for one or more diseases. However, as these models abstract several practical limitations, the realized benefits may be smaller, particularly due to a limited number of embryos and unclear future accuracy of the risk estimates. PES may negatively impact patients and their future children, as well as society. The main personal harms are an unindicated IVF treatment, a possible reduction in IVF success rates, and patient confusion, incomplete counselling, and choice overload. The main possible societal harms include discarded embryos, an increasing demand for 'designer babies', overemphasis of the genetic determinants of disease, unequal access, and lower utility in people of non-European ancestries. Benefits and harms will vary across the main potential patient groups, comprising patients already requiring IVF, fertile people with a history of a severe polygenic disease, and fertile healthy people. In the United States, the attitudes of IVF patients and the public towards PES seem positive, while healthcare professionals are cautious, sceptical about clinical utility, and concerned about patient counselling. WIDER IMPLICATIONS The theoretical potential of PES to reduce risk across multiple polygenic diseases requires further research into its benefits and harms. Given the large number of practical limitations and possible harms, particularly unnecessary IVF treatments and discarded viable embryos, PES should be offered only within a research context before further clarity is achieved regarding its balance of benefits and harms. The gap in attitudes between healthcare professionals and the public needs to be narrowed by expanding public and patient education and providing resources for informative and unbiased genetic counselling.
Collapse
Affiliation(s)
- Antonio Capalbo
- Juno Genetics, Department of Reproductive Genetics, Rome, Italy
- Center for Advanced Studies and Technology (CAST), Department of Medical Genetics, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Guido de Wert
- Department of Health, Ethics & Society, CAPHRI-School for Public Health and Primary Care and GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Heidi Mertes
- Department of Philosophy and Moral Sciences, Ghent University, Ghent, Belgium
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Liraz Klausner
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Edith Coonen
- Departments of Clinical Genetics and Reproductive Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
- School for Oncology and Developmental Biology, GROW, Maastricht University, Maastricht, The Netherlands
| | - Francesca Spinella
- Eurofins GENOMA Group Srl, Molecular Genetics Laboratories, Department of Scientific Communication, Rome, Italy
| | - Hilde Van de Velde
- Research Group Genetics Reproduction and Development (GRAD), Vrije Universiteit Brussel, Brussel, Belgium
- Brussels IVF, UZ Brussel, Brussel, Belgium
| | - Stephane Viville
- Laboratoire de Génétique Médicale LGM, Institut de Génétique Médicale d’Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, France
- Laboratoire de Diagnostic Génétique, Unité de Génétique de l’infertilité (UF3472), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Karen Sermon
- Research Group Genetics Reproduction and Development (GRAD), Vrije Universiteit Brussel, Brussel, Belgium
| | | | - Todd Lencz
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Departments of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Shai Carmi
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
27
|
Deng D, Wang H, Han K, Tang Z, Li X, Liu X, Liu X, Li X, Yu M. A genome-wide association study reveals candidate genes and regulatory regions associated with birth weight in pigs. Anim Genet 2024; 55:761-765. [PMID: 39136303 DOI: 10.1111/age.13468] [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: 06/18/2023] [Revised: 06/15/2024] [Accepted: 07/31/2024] [Indexed: 10/23/2024]
Abstract
Piglet birth weight is associated with preweaning survival, and its related traits have been included in the breeding program. Thus, understanding its genetic basis is essential. This study identified four birth weight-associated genomic regions on chromosomes 2, 4, 5, and 7 through genome-wide association study analysis in 7286 pigs from three different pure breeds using the FarmCPU model. The genetic and phenotypic variance explained by the four candidate regions is 8.42% and 1.85%, respectively. Twenty-eight candidate genes were detected, of which APPL2, TGFBI, MACROH2A1, and SEC22B have been reported to affect body growth or development. In addition, 21 H3K4me3-enriched peaks overlapped with the birth weight-associated genomic regions were identified by integrating the genome-wide association study results with our previous ChIP-seq and RNA-seq data generated in the pig placenta, a fetal organ relevant to birth weight, and three of the regulatory regions influence TGFBI, MACROH2A1, and SEC22B expression. This study provides new insights into understanding the mechanisms for birth weight. Further investigating the variants in the regulatory regions would help identify the functional variants for birth weight in pigs.
Collapse
Affiliation(s)
- Dadong Deng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hongtao Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Kun Han
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhenshuang Tang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaoping Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiangdong Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiaolei Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
| | - Xinyun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
- Hubei Hongshan Laboratory, Wuhan, China
| | - Mei Yu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
28
|
Ellis CA, Oliver KL, Harris RV, Ottman R, Scheffer IE, Mefford HC, Epstein MP, Berkovic SF, Bahlo M. Inflation of polygenic risk scores caused by sample overlap and relatedness: Examples of a major risk of bias. Am J Hum Genet 2024; 111:1805-1809. [PMID: 39168121 PMCID: PMC11393675 DOI: 10.1016/j.ajhg.2024.07.014] [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] [Received: 04/24/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Polygenic risk scores (PRSs) are an important tool for understanding the role of common genetic variants in human disease. Standard best practices recommend that PRSs be analyzed in cohorts that are independent of the genome-wide association study (GWAS) used to derive the scores without sample overlap or relatedness between the two cohorts. However, identifying sample overlap and relatedness can be challenging in an era of GWASs performed by large biobanks and international research consortia. Although most genomics researchers are aware of best practices and theoretical concerns about sample overlap and relatedness between GWAS and PRS cohorts, the prevailing assumption is that the risk of bias is small for very large GWASs. Here, we present two real-world examples demonstrating that sample overlap and relatedness is not a minor or theoretical concern but an important potential source of bias in PRS studies. Using a recently developed statistical adjustment tool, we found that excluding overlapping and related samples was equal to or more powerful than adjusting for overlap bias. Our goal is to make genomics researchers aware of the magnitude of risk of bias from sample overlap and relatedness and to highlight the need for mitigation tools, including independent validation cohorts in PRS studies, continued development of statistical adjustment methods, and tools for researchers to test their cohorts for overlap and relatedness with GWAS cohorts without sharing individual-level data.
Collapse
Affiliation(s)
- Colin A Ellis
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Karen L Oliver
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia; Population Health and Immunity Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Rebekah V Harris
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia
| | - Ruth Ottman
- Departments of Neurology and Epidemiology, and the Gertrude H. Sergievsky Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Division of Translational Epidemiology and Mental Health Equity, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Ingrid E Scheffer
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia; Department of Paediatrics, Royal Children's Hospital, University of Melbourne, Parkville, VIC 3052, Australia; The Florey Institute and Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
| | - Heather C Mefford
- Center for Pediatric Neurological Disease Research, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30307, USA
| | - Samuel F Berkovic
- Epilepsy Research Centre, Department of Medicine, University of Melbourne, Austin Health, Heidelberg, VIC 3084, Australia
| | - Melanie Bahlo
- Population Health and Immunity Division, the Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Melbourne, VIC 3052, Australia.
| |
Collapse
|
29
|
Plomin R. Nonshared environment: Real but random. JCPP ADVANCES 2024; 4:e12229. [PMID: 39411468 PMCID: PMC11472802 DOI: 10.1002/jcv2.12229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 11/13/2023] [Indexed: 10/19/2024] Open
Abstract
Background In the excitement about genomics, it is easy to lose sight of one of the most important findings from behavioural genetics: At least half of the variance of psychopathology is caused by environmental effects that are not shared by children growing up in the same family, which includes error of measurement. However, a 30-year search for the systematic causes of nonshared environment in a line-up of the usual suspects, especially parenting, has not identified the culprits. Method I briefly review this research, but primarily consider the conceptual framework of the search for 'missing' nonshared environmental effects. Results The search has focused on exogenous events like parenting, but nonshared environment might not be caused by anything we would call an event. Instead, it might reflect endogenous processes such as noisy biological systems (such as somatic mutations and epigenetics) or, at a psychological level, idiosyncratic subjective perceptions of past and present experiences, which could be called nonshared environmental experience to distinguish it from exogenous events. Although real, nonshared environment might be random in the philosophy of science sense of being unpredictable, even though it can have stable effects that predict subsequent behaviour. Conclusion I wade into the weeds of randomness and suggest that this so-called 'gloomy prospect' might not be so gloomy.
Collapse
Affiliation(s)
- Robert Plomin
- King's College LondonInstitute of Psychiatry, Psychology and NeuroscienceLondonUK
| |
Collapse
|
30
|
Sullivan PF, Yao S, Hjerling-Leffler J. Schizophrenia genomics: genetic complexity and functional insights. Nat Rev Neurosci 2024; 25:611-624. [PMID: 39030273 DOI: 10.1038/s41583-024-00837-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2024] [Indexed: 07/21/2024]
Abstract
Determining the causes of schizophrenia has been a notoriously intractable problem, resistant to a multitude of investigative approaches over centuries. In recent decades, genomic studies have delivered hundreds of robust findings that implicate nearly 300 common genetic variants (via genome-wide association studies) and more than 20 rare variants (via whole-exome sequencing and copy number variant studies) as risk factors for schizophrenia. In parallel, functional genomic and neurobiological studies have provided exceptionally detailed information about the cellular composition of the brain and its interconnections in neurotypical individuals and, increasingly, in those with schizophrenia. Taken together, these results suggest unexpected complexity in the mechanisms that drive schizophrenia, pointing to the involvement of ensembles of genes (polygenicity) rather than single-gene causation. In this Review, we describe what we now know about the genetics of schizophrenia and consider the neurobiological implications of this information.
Collapse
Affiliation(s)
- Patrick F Sullivan
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
- Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| | - Shuyang Yao
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jens Hjerling-Leffler
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
31
|
Cook SR, Hugen S, Hayward JJ, Famula TR, Belanger JM, McNiel E, Fieten H, Oberbauer AM, Leegwater PA, Ostrander EA, Mandigers PJ, Evans JM. Genomic analyses identify 15 susceptibility loci and reveal HDAC2, SOX2-OT, and IGF2BP2 in a naturally-occurring canine model of gastric cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.604426. [PMID: 39372775 PMCID: PMC11451740 DOI: 10.1101/2024.08.14.604426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Gastric cancer (GC) is the fifth most common human cancer worldwide, but the genetic etiology is largely unknown. We performed a Bayesian genome-wide association study and selection analyses in a naturally-occurring canine model of GC, the Belgian Tervuren and Sheepdog breeds, to elucidate underlying genetic risk factors. We identified 15 loci with over 90% predictive accuracy for the GC phenotype. Variant filtering revealed germline putative regulatory variants for the EPAS1 (HIF2A) and PTEN genes and a coding variant in CD101. Although closely related to Tervuren and Sheepdogs, Belgian Malinois rarely develop GC. Across-breed analyses uncovered protective haplotypes under selection in Malinois at SOX2-OT and IGF2BP2. Among Tervuren and Sheepdogs, HDAC2 putative regulatory variants were present at comparatively high frequency and were associated with GC. Here, we describe a complex genetic architecture governing GC in a dog model, including genes such as PDZRN3, that have not been associated with human GC.
Collapse
Affiliation(s)
- Shawna R. Cook
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Sanne Hugen
- Expertisecentre of Genetics, Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jessica J. Hayward
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Thomas R. Famula
- Department of Animal Science, University of California, Davis, CA, USA
| | | | - Elizabeth McNiel
- Cummings School of Veterinary Medicine, Tufts University, Grafton, Massachusetts, USA
| | - Hille Fieten
- Expertisecentre of Genetics, Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - Peter A.J. Leegwater
- Expertisecentre of Genetics, Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Elaine A. Ostrander
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Center, National Institutes of Health, Bethesda, MD, USA
| | - Paul J.J. Mandigers
- Expertisecentre of Genetics, Department of Clinical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jacquelyn M. Evans
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| |
Collapse
|
32
|
Don J, Schork AJ, Glusman G, Rappaport N, Cummings SR, Duggan D, Raju A, Hellberg KLG, Gunn S, Monti S, Perls T, Lapidus J, Goetz LH, Sebastiani P, Schork NJ. The relationship between 11 different polygenic longevity scores, parental lifespan, and disease diagnosis in the UK Biobank. GeroScience 2024; 46:3911-3927. [PMID: 38451433 PMCID: PMC11226417 DOI: 10.1007/s11357-024-01107-1] [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: 10/30/2023] [Accepted: 02/21/2024] [Indexed: 03/08/2024] Open
Abstract
Large-scale genome-wide association studies (GWAS) strongly suggest that most traits and diseases have a polygenic component. This observation has motivated the development of disease-specific "polygenic scores (PGS)" that are weighted sums of the effects of disease-associated variants identified from GWAS that correlate with an individual's likelihood of expressing a specific phenotype. Although most GWAS have been pursued on disease traits, leading to the creation of refined "Polygenic Risk Scores" (PRS) that quantify risk to diseases, many GWAS have also been pursued on extreme human longevity, general fitness, health span, and other health-positive traits. These GWAS have discovered many genetic variants seemingly protective from disease and are often different from disease-associated variants (i.e., they are not just alternative alleles at disease-associated loci) and suggest that many health-positive traits also have a polygenic basis. This observation has led to an interest in "polygenic longevity scores (PLS)" that quantify the "risk" or genetic predisposition of an individual towards health. We derived 11 different PLS from 4 different available GWAS on lifespan and then investigated the properties of these PLS using data from the UK Biobank (UKB). Tests of association between the PLS and population structure, parental lifespan, and several cancerous and non-cancerous diseases, including death from COVID-19, were performed. Based on the results of our analyses, we argue that PLS are made up of variants not only robustly associated with parental lifespan, but that also contribute to the genetic architecture of disease susceptibility, morbidity, and mortality.
Collapse
Affiliation(s)
- Janith Don
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Andrew J Schork
- The Institute of Biological Psychiatry, Copenhagen University Hospital, Copenhagen, Denmark
- GLOBE Institute, Copenhagen University, Copenhagen, Denmark
| | | | | | - Steve R Cummings
- San Francisco Coordinating Center, California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - David Duggan
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Anish Raju
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Kajsa-Lotta Georgii Hellberg
- The Institute of Biological Psychiatry, Copenhagen University Hospital, Copenhagen, Denmark
- GLOBE Institute, Copenhagen University, Copenhagen, Denmark
| | - Sophia Gunn
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Stefano Monti
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Thomas Perls
- Department of Medicine, Section of Geriatrics, Boston University, Boston, MA, USA
| | - Jodi Lapidus
- Department of Biostatistics, Oregon Health & Science University, Portland, OR, USA
| | - Laura H Goetz
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
- Veterans Affairs Loma Linda Health Care, Loma Linda, CA, USA
| | - Paola Sebastiani
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
- Tufts University School of Medicine and Data Intensive Study Center, Boston, MA, USA
| | - Nicholas J Schork
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
- The City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
33
|
Treccani M, Veschetti L, Patuzzo C, Malerba G, Vaglio A, Martorana D. Genetic and Non-Genetic Contributions to Eosinophilic Granulomatosis with Polyangiitis: Current Knowledge and Future Perspectives. Curr Issues Mol Biol 2024; 46:7516-7529. [PMID: 39057087 PMCID: PMC11275403 DOI: 10.3390/cimb46070446] [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: 05/31/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
In this work, we present a comprehensive overview of the genetic and non-genetic complexity of eosinophilic granulomatosis with polyangiitis (EGPA). EGPA is a rare complex systemic disease that occurs in people presenting with severe asthma and high eosinophilia. After briefly introducing EGPA and its relationship with the anti-neutrophil cytoplasmic autoantibodies (ANCA)-associated vasculitis (AAVs), we delve into the complexity of this disease. At first, the two main biological actors, ANCA and eosinophils, are presented. Biological and clinical phenotypes related to ANCA positivity or negativity are explained, as well as the role of eosinophils and their pathological subtypes, pointing out their intricate relations with EGPA. Then, the genetics of EGPA are described, providing an overview of the research effort to unravel them. Candidate gene studies have investigated biologically relevant candidate genes; the more recent genome-wide association studies and meta-analyses, able to analyze the whole genome, have confirmed previous associations and discovered novel risk loci; in the end, family-based studies have dissected the contribution of rare variants and the heritability of EGPA. Then, we briefly present the environmental contribution to EGPA, reporting seasonal events and pollutants as triggering factors. In the end, the latest omic research is discussed and the most recent epigenomic, transcriptomic and microbiome studies are presented, highlighting the current challenges, open questions and suggesting approaches to unraveling this complex disease.
Collapse
Affiliation(s)
- Mirko Treccani
- GM Lab, Department of Surgery, Dentistry, Gynaecology and Paediatrics, University of Verona, 37134 Verona, Italy;
| | - Laura Veschetti
- Infections and Cystic Fibrosis Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milano, Italy;
- Vita-Salute San Raffaele University, 20132 Milano, Italy
| | - Cristina Patuzzo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy;
| | - Giovanni Malerba
- GM Lab, Department of Surgery, Dentistry, Gynaecology and Paediatrics, University of Verona, 37134 Verona, Italy;
| | - Augusto Vaglio
- Nephrology and Dialysis Unit, Meyer Children’s Hospital IRCCS, 50139 Florence, Italy;
- Department of Biomedical Experimental and Clinical Sciences “Mario Serio”, University of Florence, 50121 Florence, Italy
| | - Davide Martorana
- Medical Genetics Unit, Department of Onco-Hematology, University Hospital of Parma, 43126 Parma, Italy;
- CoreLab Unit, Research Center, University Hospital of Parma, 43126 Parma, Italy
| |
Collapse
|
34
|
Chen Y, Wang G, Chen J, Wang C, Dong X, Chang HM, Yuan S, Zhao Y, Mu L. Genetic and Epigenetic Landscape for Drug Development in Polycystic Ovary Syndrome. Endocr Rev 2024; 45:437-459. [PMID: 38298137 DOI: 10.1210/endrev/bnae002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/26/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024]
Abstract
The treatment of polycystic ovary syndrome (PCOS) faces challenges as all known treatments are merely symptomatic. The US Food and Drug Administration has not approved any drug specifically for treating PCOS. As the significance of genetics and epigenetics rises in drug development, their pivotal insights have greatly enhanced the efficacy and success of drug target discovery and validation, offering promise for guiding the advancement of PCOS treatments. In this context, we outline the genetic and epigenetic advancement in PCOS, which provide novel insights into the pathogenesis of this complex disease. We also delve into the prospective method for harnessing genetic and epigenetic strategies to identify potential drug targets and ensure target safety. Additionally, we shed light on the preliminary evidence and distinctive challenges associated with gene and epigenetic therapies in the context of PCOS.
Collapse
Affiliation(s)
- Yi Chen
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Guiquan Wang
- Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen 361003, China
- Xiamen Key Laboratory of Reproduction and Genetics, Xiamen University, Xiamen 361023, China
| | - Jingqiao Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou 325035, China
| | - Congying Wang
- The Department of Cardiology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang 322000, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung 40400, Taiwan
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm 171 65, Sweden
| | - Yue Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing 100007, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University, Beijing 100191, China
| | - Liangshan Mu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
35
|
Benstock SE, Weaver K, Hettema JM, Verhulst B. Using Alternative Definitions of Controls to Increase Statistical Power in GWAS. Behav Genet 2024; 54:353-366. [PMID: 38869698 PMCID: PMC11661655 DOI: 10.1007/s10519-024-10187-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024]
Abstract
Genome-wide association studies (GWAS) are often underpowered due to small effect sizes of common single nucleotide polymorphisms (SNPs) on phenotypes and extreme multiple testing thresholds. The most common approach for increasing statistical power is to increase sample size. We propose an alternative strategy of redefining case-control outcomes into ordinal case-subthreshold-asymptomatic variables. While maintaining the clinical case threshold, we subdivide controls into two groups: individuals who are symptomatic but do not meet the clinical criteria for diagnosis (subthreshold) and individuals who are effectively asymptomatic. We conducted a simulation study to examine the impact of effect size, minor allele frequency, population prevalence, and the prevalence of the subthreshold group on statistical power to detect genetic associations in three scenarios: a standard case-control, an ordinal, and a case-asymptomatic control analysis. Our results suggest the ordinal model consistently provides the greatest statistical power while the case-control model the least. Power in the case-asymptomatic control model reflects the case-control or ordinal model depending on the population prevalence and size of the subthreshold category. We then analyzed a major depression phenotype from the UK Biobank to corroborate our simulation results. Overall, the ordinal model improves statistical power in GWAS consistent with increasing the sample size by approximately 10%.
Collapse
Affiliation(s)
- Sarah E Benstock
- Department of Psychiatry and Behavioral Sciences, Texas A&M University School of Medicine, College Station, TX, USA
| | - Katherine Weaver
- Department of Psychiatry and Behavioral Sciences, Texas A&M University School of Medicine, College Station, TX, USA
| | - John M Hettema
- Department of Psychiatry and Behavioral Sciences, Texas A&M University School of Medicine, College Station, TX, USA
| | - Brad Verhulst
- Department of Psychiatry and Behavioral Sciences, Texas A&M University School of Medicine, College Station, TX, USA.
| |
Collapse
|
36
|
Sasner M, Preuss C, Pandey RS, Uyar A, Garceau D, Kotredes KP, Williams H, Oblak AL, Lin PB, Perkins B, Soni D, Ingraham C, Lee‐Gosselin A, Lamb BT, Howell GR, Carter GW. In vivo validation of late-onset Alzheimer's disease genetic risk factors. Alzheimers Dement 2024; 20:4970-4984. [PMID: 38687251 PMCID: PMC11247676 DOI: 10.1002/alz.13840] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Genome-wide association studies have identified over 70 genetic loci associated with late-onset Alzheimer's disease (LOAD), but few candidate polymorphisms have been functionally assessed for disease relevance and mechanism of action. METHODS Candidate genetic risk variants were informatically prioritized and individually engineered into a LOAD-sensitized mouse model that carries the AD risk variants APOE ε4/ε4 and Trem2*R47H. The potential disease relevance of each model was assessed by comparing brain transcriptomes measured with the Nanostring Mouse AD Panel at 4 and 12 months of age with human study cohorts. RESULTS We created new models for 11 coding and loss-of-function risk variants. Transcriptomic effects from multiple genetic variants recapitulated a variety of human gene expression patterns observed in LOAD study cohorts. Specific models matched to emerging molecular LOAD subtypes. DISCUSSION These results provide an initial functionalization of 11 candidate risk variants and identify potential preclinical models for testing targeted therapeutics. HIGHLIGHTS A novel approach to validate genetic risk factors for late-onset AD (LOAD) is presented. LOAD risk variants were knocked in to conserved mouse loci. Variant effects were assayed by transcriptional analysis. Risk variants in Abca7, Mthfr, Plcg2, and Sorl1 loci modeled molecular signatures of clinical disease. This approach should generate more translationally relevant animal models.
Collapse
Affiliation(s)
| | | | - Ravi S. Pandey
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Asli Uyar
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | | | | | | | - Adrian L. Oblak
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Peter Bor‐Chian Lin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bridget Perkins
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Disha Soni
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Cindy Ingraham
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Audrey Lee‐Gosselin
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | - Bruce T. Lamb
- Stark Neurosciences Research Institute, School of Medicine, Indiana UniversityIndianapolisIndianaUSA
| | | | - Gregory W. Carter
- The Jackson LaboratoryBar HarborMaineUSA
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| |
Collapse
|
37
|
Puga M, Serrano JG, García EL, González Carracedo MA, Jiménez-Canino R, Pino-Yanes M, Karlsson R, Sullivan PF, Fregel R. El Hierro Genome Study: A Genomic and Health Study in an Isolated Canary Island Population. J Pers Med 2024; 14:626. [PMID: 38929847 PMCID: PMC11204744 DOI: 10.3390/jpm14060626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
El Hierro is the smallest and westernmost island of the Canary Islands, whose population derives from an admixture of different ancestral components and that has been subjected to genetic isolation. We established the "El Hierro Genome Study" to characterize the health status and the genetic composition of ~10% of the current population of the island, accounting for a total of 1054 participants. Detailed demographic and clinical data and a blood sample for DNA extraction were obtained from each participant. Genomic genotyping was performed with the Global Screening Array (Illumina). The genetic composition of El Hierro was analyzed in a subset of 416 unrelated individuals by characterizing the mitochondrial DNA (mtDNA) and Y-chromosome haplogroups and performing principal component analyses (PCAs). In order to explore signatures of isolation, runs of homozygosity (ROHs) were also estimated. Among the participants, high blood pressure, hypercholesterolemia, and diabetes were the most prevalent conditions. The most common mtDNA haplogroups observed were of North African indigenous origin, while the Y-chromosome ones were mainly European. The PCA showed that the El Hierro population clusters near 1000 Genomes' European population but with a shift toward African populations. Moreover, the ROH analysis revealed some individuals with an important portion of their genomes with ROHs exceeding 400 Mb. Overall, these results confirmed that the "El Hierro Genome" cohort offers an opportunity to study the genetic basis of several diseases in an unexplored isolated population.
Collapse
Affiliation(s)
- Marta Puga
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (M.P.); (E.L.G.); (M.A.G.C.); (M.P.-Y.)
| | - Javier G. Serrano
- Evolution, Paleogenomics and Population Genetics Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), 38200 La Laguna, Spain;
| | - Elsa L. García
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (M.P.); (E.L.G.); (M.A.G.C.); (M.P.-Y.)
| | - Mario A. González Carracedo
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (M.P.); (E.L.G.); (M.A.G.C.); (M.P.-Y.)
- Genetics Laboratory, Institute of Tropical Diseases and Public Health of the Canary Islands (IUETSPC), Universidad de La Laguna (ULL), 38200 La Laguna, Spain
| | - Rubén Jiménez-Canino
- Genomics Service, Servicio General de Apoyo a la Investigación, Universidad de La Laguna (ULL), 38200 La Laguna, Spain;
| | - María Pino-Yanes
- Genomics and Health Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), 38200 La Laguna, Spain; (M.P.); (E.L.G.); (M.A.G.C.); (M.P.-Y.)
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Instituto de Tecnologías Biomédicas (ITB), Universidad de La Laguna (ULL), 38200 La Laguna, Spain
| | - Robert Karlsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden; (R.K.); (P.F.S.)
| | - Patrick F. Sullivan
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77 Stockholm, Sweden; (R.K.); (P.F.S.)
- Departments of Genetics and Psychiatry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rosa Fregel
- Evolution, Paleogenomics and Population Genetics Group, Department of Biochemistry, Microbiology, Cell Biology and Genetics, Universidad de La Laguna (ULL), 38200 La Laguna, Spain;
| |
Collapse
|
38
|
Gualdi F, Oliva B, Piñero J. Predicting gene disease associations with knowledge graph embeddings for diseases with curtailed information. NAR Genom Bioinform 2024; 6:lqae049. [PMID: 38745993 PMCID: PMC11091931 DOI: 10.1093/nargab/lqae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/08/2024] [Accepted: 04/24/2024] [Indexed: 05/16/2024] Open
Abstract
Knowledge graph embeddings (KGE) are a powerful technique used in the biomedical domain to represent biological knowledge in a low dimensional space. However, a deep understanding of these methods is still missing, and, in particular, regarding their applications to prioritize genes associated with complex diseases with reduced genetic information. In this contribution, we built a knowledge graph (KG) by integrating heterogeneous biomedical data and generated KGE by implementing state-of-the-art methods, and two novel algorithms: Dlemb and BioKG2vec. Extensive testing of the embeddings with unsupervised clustering and supervised methods showed that KGE can be successfully implemented to predict genes associated with diseases and that our novel approaches outperform most existing algorithms in both scenarios. Our findings underscore the significance of data quality, preprocessing, and integration in achieving accurate predictions. Additionally, we applied KGE to predict genes linked to Intervertebral Disc Degeneration (IDD) and illustrated that functions pertinent to the disease are enriched within the prioritized gene set.
Collapse
Affiliation(s)
- Francesco Gualdi
- Integrative Biomedical Informatics, Research Programme on Biomedical Informatics (IBI-GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain
- Structural Bioinformatics Lab, Research Programme on Biomedical Informatics (SBI-GRIB), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain
| | - Baldomero Oliva
- Structural Bioinformatics Lab, Research Programme on Biomedical Informatics (SBI-GRIB), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain
| | - Janet Piñero
- Integrative Biomedical Informatics, Research Programme on Biomedical Informatics (IBI-GRIB), Hospital del Mar Medical Research Institute (IMIM), Department of Experimental and Health Sciences, Universitat Pompeu Fabra, C/Dr Aiguader 88, E-08003 Barcelona, Spain
- Medbioinformatics Solutions SL, Barcelona, Spain
| |
Collapse
|
39
|
Bicks LK, Geschwind DH. Functional neurogenomics in autism spectrum disorders: A decade of progress. Curr Opin Neurobiol 2024; 86:102858. [PMID: 38547564 DOI: 10.1016/j.conb.2024.102858] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 06/11/2024]
Abstract
Advances in autism spectrum disorder (ASD) genetics have identified many genetic causes, reflecting remarkable progress while at the same time identifying challenges such as heterogeneity and pleiotropy, which complicate attempts to connect genetic risk to mechanisms. High-throughput functional genomic approaches have yielded progress by defining a molecular pathology in the brain of individuals with ASD and in identifying convergent biological pathways through which risk genes are predicted to act. These studies indicate that ASD genetic risk converges in early brain development, primarily during the period of cortical neurogenesis. Over development, genetic perturbations in turn lead to broad neuronal signaling dysregulation, most prominent in glutamatergic cortical-cortical projecting neurons and somatostatin positive interneurons, which is accompanied by glial dyshomeostasis throughout the cerebral cortex. Connecting these developmental perturbations to disrupted neuronal and glial function in the postnatal brain is an important direction in current research. Coupling functional genomic approaches with advances in induced pluripotent stem cell-derived neural organoid development provides a promising avenue for connecting brain pathology to developmental mechanisms.
Collapse
Affiliation(s)
- Lucy K Bicks
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA. https://twitter.com/Bickslucy
| | - D H Geschwind
- Program in Neurobehavioral Genetics, Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Department of Psychiatry, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, 695 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
40
|
Randolph HE, Aracena KA, Lin YL, Mu Z, Barreiro LB. Shaping immunity: The influence of natural selection on population immune diversity. Immunol Rev 2024; 323:227-240. [PMID: 38577999 DOI: 10.1111/imr.13329] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
Humans exhibit considerable variability in their immune responses to the same immune challenges. Such variation is widespread and affects individual and population-level susceptibility to infectious diseases and immune disorders. Although the factors influencing immune response diversity are partially understood, what mechanisms lead to the wide range of immune traits in healthy individuals remain largely unexplained. Here, we discuss the role that natural selection has played in driving phenotypic differences in immune responses across populations and present-day susceptibility to immune-related disorders. Further, we touch on future directions in the field of immunogenomics, highlighting the value of expanding this work to human populations globally, the utility of modeling the immune response as a dynamic process, and the importance of considering the potential polygenic nature of natural selection. Identifying loci acted upon by evolution may further pinpoint variants critically involved in disease etiology, and designing studies to capture these effects will enrich our understanding of the genetic contributions to immunity and immune dysregulation.
Collapse
Affiliation(s)
- Haley E Randolph
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | | | - Yen-Lung Lin
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Zepeng Mu
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, Illinois, USA
| | - Luis B Barreiro
- Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, Illinois, USA
- Department of Human Genetics, University of Chicago, Chicago, Illinois, USA
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
41
|
Zhang J, Chen W, Chen G, Flannick J, Fikse E, Smerin G, Degner K, Yang Y, Xu C, Consortium AMP-T2D-GENES, Li Y, Hanover JA, Simonds WF. Ancestry-specific high-risk gene variant profiling unmasks diabetes-associated genes. Hum Mol Genet 2024; 33:655-666. [PMID: 36255737 PMCID: PMC11000659 DOI: 10.1093/hmg/ddac255] [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] [Received: 05/04/2022] [Revised: 09/28/2022] [Accepted: 10/10/2022] [Indexed: 11/15/2022] Open
Abstract
How ancestry-associated genetic variance affects disparities in the risk of polygenic diseases and influences the identification of disease-associated genes warrants a deeper understanding. We hypothesized that the discovery of genes associated with polygenic diseases may be limited by the overreliance on single-nucleotide polymorphism (SNP)-based genomic investigation, as most significant variants identified in genome-wide SNP association studies map to introns and intergenic regions of the genome. To overcome such potential limitations, we developed a gene-constrained, function-based analytical method centered on high-risk variants (hrV) that encode frameshifts, stopgains or splice site disruption. We analyzed the total number of hrV per gene in populations of different ancestry, representing a total of 185 934 subjects. Using this analysis, we developed a quantitative index of hrV (hrVI) across 20 428 genes within each population. We then applied hrVI analysis to the discovery of genes associated with type 2 diabetes mellitus (T2DM), a polygenic disease with ancestry-related disparity. HrVI profiling and gene-to-gene comparisons of ancestry-specific hrV between the case (20 781 subjects) and control (24 440 subjects) populations in the T2DM national repository identified 57 genes associated with T2DM, 40 of which were discoverable only by ancestry-specific analysis. These results illustrate how a function-based, ancestry-specific analysis of genetic variations can accelerate the identification of genes associated with polygenic diseases. Besides T2DM, such analysis may facilitate our understanding of the genetic basis for other polygenic diseases that are also greatly influenced by environmental and behavioral factors, such as obesity, hypertension and Alzheimer's disease.
Collapse
Affiliation(s)
- Jianhua Zhang
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
| | - Weiping Chen
- Genomic Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
| | - Guanjie Chen
- Center for Research on Genomics and Global Health, National Human Genome Research Institute, Bethesda, MD 20892, United States
| | - Jason Flannick
- Metabolism Program, Broad Institute, Cambridge, MA 02142, United States
| | - Emma Fikse
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
| | - Glenda Smerin
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
| | - Katherine Degner
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
| | - Yanqin Yang
- Laboratory of Transplantation Genomics, National Heart Lung and Blood Institute; National Institutes of Health, Bethesda, MD 20892, United States
| | - Catherine Xu
- Genomic Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
| | | | - Yulong Li
- Milton S. Hershey Medical Center, Division of Endocrinology, Diabetes and Metabolism, Penn State University, Hershey, PA 17033, United States
| | - John A Hanover
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
| | - William F Simonds
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, United States
| |
Collapse
|
42
|
Odriozola A, González A, Álvarez-Herms J, Corbi F. Sleep regulation and host genetics. ADVANCES IN GENETICS 2024; 111:497-535. [PMID: 38908905 DOI: 10.1016/bs.adgen.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Due to the multifactorial and complex nature of rest, we focus on phenotypes related to sleep. Sleep regulation is a multifactorial process. In this chapter, we focus on those phenotypes inherent to sleep that are highly prevalent in the population, and that can be modulated by lifestyle, such as sleep quality and duration, insomnia, restless leg syndrome and daytime sleepiness. We, therefore, leave in the background those phenotypes that constitute infrequent pathologies or for which the current level of scientific evidence does not favour the implementation of practical approaches of this type. Similarly, the regulation of sleep quality is intimately linked to the regulation of the circadian rhythm. Although this relationship is discussed in the sections that require it, the in-depth study of circadian rhythm regulation at the molecular level deserves a separate chapter, and this is how it is dealt with in this volume.
Collapse
Affiliation(s)
- Adrián Odriozola
- Hologenomiks Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Adriana González
- Hologenomiks Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jesús Álvarez-Herms
- Phymo® Lab, Physiology, and Molecular Laboratory, Collado Hermoso, Segovia, Spain
| | - Francesc Corbi
- Institut Nacional d'Educació Física de Catalunya (INEFC), Centre de Lleida, Universitat de Lleida (UdL), Lleida, Spain
| |
Collapse
|
43
|
Yurkovich JT, Evans SJ, Rappaport N, Boore JL, Lovejoy JC, Price ND, Hood LE. The transition from genomics to phenomics in personalized population health. Nat Rev Genet 2024; 25:286-302. [PMID: 38093095 DOI: 10.1038/s41576-023-00674-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2023] [Indexed: 03/21/2024]
Abstract
Modern health care faces several serious challenges, including an ageing population and its inherent burden of chronic diseases, rising costs and marginal quality metrics. By assessing and optimizing the health trajectory of each individual using a data-driven personalized approach that reflects their genetics, behaviour and environment, we can start to address these challenges. This assessment includes longitudinal phenome measures, such as the blood proteome and metabolome, gut microbiome composition and function, and lifestyle and behaviour through wearables and questionnaires. Here, we review ongoing large-scale genomics and longitudinal phenomics efforts and the powerful insights they provide into wellness. We describe our vision for the transformation of the current health care from disease-oriented to data-driven, wellness-oriented and personalized population health.
Collapse
Affiliation(s)
- James T Yurkovich
- Phenome Health, Seattle, WA, USA
- Center for Phenomic Health, The Buck Institute for Research on Aging, Novato, CA, USA
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, USA
| | - Simon J Evans
- Phenome Health, Seattle, WA, USA
- Center for Phenomic Health, The Buck Institute for Research on Aging, Novato, CA, USA
| | - Noa Rappaport
- Center for Phenomic Health, The Buck Institute for Research on Aging, Novato, CA, USA
- Institute for Systems Biology, Seattle, WA, USA
| | - Jeffrey L Boore
- Phenome Health, Seattle, WA, USA
- Center for Phenomic Health, The Buck Institute for Research on Aging, Novato, CA, USA
| | - Jennifer C Lovejoy
- Phenome Health, Seattle, WA, USA
- Center for Phenomic Health, The Buck Institute for Research on Aging, Novato, CA, USA
- Institute for Systems Biology, Seattle, WA, USA
| | - Nathan D Price
- Institute for Systems Biology, Seattle, WA, USA
- Thorne HealthTech, New York, NY, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| | - Leroy E Hood
- Phenome Health, Seattle, WA, USA.
- Center for Phenomic Health, The Buck Institute for Research on Aging, Novato, CA, USA.
- Institute for Systems Biology, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA.
- Department of Immunology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
44
|
Zhang J, Pandey M, Awe A, Lue N, Kittock C, Fikse E, Degner K, Staples J, Mokhasi N, Chen W, Yang Y, Adikaram P, Jacob N, Greenfest-Allen E, Thomas R, Bomeny L, Zhang Y, Petros TJ, Wang X, Li Y, Simonds WF. The association of GNB5 with Alzheimer disease revealed by genomic analysis restricted to variants impacting gene function. Am J Hum Genet 2024; 111:473-486. [PMID: 38354736 PMCID: PMC10940018 DOI: 10.1016/j.ajhg.2024.01.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 02/16/2024] Open
Abstract
Disease-associated variants identified from genome-wide association studies (GWASs) frequently map to non-coding areas of the genome such as introns and intergenic regions. An exclusive reliance on gene-agnostic methods of genomic investigation could limit the identification of relevant genes associated with polygenic diseases such as Alzheimer disease (AD). To overcome such potential restriction, we developed a gene-constrained analytical method that considers only moderate- and high-risk variants that affect gene coding sequences. We report here the application of this approach to publicly available datasets containing 181,388 individuals without and with AD and the resulting identification of 660 genes potentially linked to the higher AD prevalence among Africans/African Americans. By integration with transcriptome analysis of 23 brain regions from 2,728 AD case-control samples, we concentrated on nine genes that potentially enhance the risk of AD: AACS, GNB5, GNS, HIPK3, MED13, SHC2, SLC22A5, VPS35, and ZNF398. GNB5, the fifth member of the heterotrimeric G protein beta family encoding Gβ5, is primarily expressed in neurons and is essential for normal neuronal development in mouse brain. Homozygous or compound heterozygous loss of function of GNB5 in humans has previously been associated with a syndrome of developmental delay, cognitive impairment, and cardiac arrhythmia. In validation experiments, we confirmed that Gnb5 heterozygosity enhanced the formation of both amyloid plaques and neurofibrillary tangles in the brains of AD model mice. These results suggest that gene-constrained analysis can complement the power of GWASs in the identification of AD-associated genes and may be more broadly applicable to other polygenic diseases.
Collapse
Affiliation(s)
- Jianhua Zhang
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Mritunjay Pandey
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Adam Awe
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicole Lue
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Claire Kittock
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emma Fikse
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine Degner
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jenna Staples
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Neha Mokhasi
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weiping Chen
- Genomic Core, National Institute of Diabetes and Digestive and Kidney Diseases, Bldg. 8/Rm 1A11, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yanqin Yang
- Laboratory of Transplantation Genomics, National Heart Lung and Blood Institute, Bldg. 10/Rm 7S261, National Institutes of Health, Bethesda, MD 20892, USA
| | - Poorni Adikaram
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nirmal Jacob
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Emily Greenfest-Allen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rachel Thomas
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laura Bomeny
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yajun Zhang
- Unit on Cellular and Molecular Neurodevelopment, Bldg. 35/Rm 3B 1002, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy J Petros
- Unit on Cellular and Molecular Neurodevelopment, Bldg. 35/Rm 3B 1002, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiaowen Wang
- Partek Incorporated, 12747 Olive Boulevard, St. Louis, MO 63141, USA
| | - Yulong Li
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA
| | - William F Simonds
- Metabolic Diseases Branch, Bldg. 10/Rm 8C-101, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
45
|
Valentim WL, Tylee DS, Polimanti R. A perspective on translating genomic discoveries into targets for brain-machine interface and deep brain stimulation devices. WIREs Mech Dis 2024; 16:e1635. [PMID: 38059513 PMCID: PMC11163995 DOI: 10.1002/wsbm.1635] [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: 03/20/2023] [Revised: 10/22/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023]
Abstract
Mental illnesses have a huge impact on individuals, families, and society, so there is a growing need for more efficient treatments. In this context, brain-computer interface (BCI) technology has the potential to revolutionize the options for neuropsychiatric therapies. However, the development of BCI-based therapies faces enormous challenges, such as power dissipation constraints, lack of credible feedback mechanisms, uncertainty of which brain areas and frequencies to target, and even which patients to treat. Some of these setbacks are due to the large gap in our understanding of brain function. In recent years, large-scale genomic analyses uncovered an unprecedented amount of information regarding the biology of the altered brain function observed across the psychopathology spectrum. We believe findings from genetic studies can be useful to refine BCI technology to develop novel treatment options for mental illnesses. Here, we assess the latest advancements in both fields, the possibilities that can be generated from their intersection, and the challenges that these research areas will need to address to ensure that translational efforts can lead to effective and reliable interventions. Specifically, starting from highlighting the overlap between mechanisms uncovered by large-scale genetic studies and the current targets of deep brain stimulation treatments, we describe the steps that could help to translate genomic discoveries into BCI targets. Because these two research areas have not been previously presented together, the present article can provide a novel perspective for scientists with different research backgrounds. This article is categorized under: Neurological Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Biomedical Engineering.
Collapse
Affiliation(s)
- Wander L. Valentim
- Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
| | - Daniel S. Tylee
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
- VA CT Healthcare Center, West Haven, CT, USA
| | - Renato Polimanti
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT
- VA CT Healthcare Center, West Haven, CT, USA
| |
Collapse
|
46
|
Kang J, Wei S, Jia Z, Ma Y, Chen H, Sun C, Xu J, Tao J, Dong Y, Lv W, Tian H, Guo X, Bi S, Zhang C, Jiang Y, Lv H, Zhang M. Effects of genetic variation on the structure of RNA and protein. Proteomics 2024; 24:e2300235. [PMID: 38197532 DOI: 10.1002/pmic.202300235] [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: 05/31/2023] [Revised: 12/15/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024]
Abstract
Changes in the structure of RNA and protein, have an important impact on biological functions and are even important determinants of disease pathogenesis and treatment. Some genetic variations, including copy number variation, single nucleotide variation, and so on, can lead to changes in biological function and increased susceptibility to certain diseases by changing the structure of RNA or protein. With the development of structural biology and sequencing technology, a large amount of RNA and protein structure data and genetic variation data resources has emerged to be used to explain biological processes. Here, we reviewed the effects of genetic variation on the structure of RNAs and proteins, and investigated their impact on several diseases. An online resource (http://www.onethird-lab.com/gems/) to support convenient retrieval of common tools is also built. Finally, the challenges and future development of the effects of genetic variation on RNA and protein were discussed.
Collapse
Affiliation(s)
- Jingxuan Kang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Siyu Wei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Zhe Jia
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Yingnan Ma
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Haiyan Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Chen Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Jing Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Junxian Tao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Yu Dong
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Wenhua Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, 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
| | - Yongshuai Jiang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Hongchao Lv
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| | - Mingming Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
- The Epigenome-Wide Association Study Project, Harbin, China
| |
Collapse
|
47
|
Rauf T, Freese J. Genetic influences on depression and selection into adverse life experiences. Soc Sci Med 2024; 344:116633. [PMID: 38324978 DOI: 10.1016/j.socscimed.2024.116633] [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] [Received: 10/17/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
Genome-wide association studies find that a large number of genetic variants jointly influence the risk of depression, which is summarized by polygenic indices (PGIs) of depressive symptoms and major depression. But PGIs by design remain agnostic about the causal mechanisms linking genes to depression. Meanwhile, the role of adverse life experiences in shaping depression risk is well-documented, including via gene-environment correlation. Building on theoretical work on dynamic and contingent genetic selection, we suggest that genetic influences may lead to differential selection into negative life experiences, forging gene-environment correlations that manifest in various permutations of depressive behaviors and environmental adversities. We also examine the extent to which apparent genetic influences may reflect spurious associations due to factors such as indirect genetic effects. Using data from two large surveys of middle-aged and older US adults, we investigate to what extent a PGI of depression predicts the risk of 27 different adversities. Further, to glean insights about the kinds of processes that might lead to gene-environment correlation, we augment these analyses with data from an original preregistered survey to measure cultural understandings of the behavioral dependence of various adversities. We find that the PGI predicts the risk of majority of adversities, net of class background and prior depression, and that the selection risk is greater for adversities typically perceived as being dependent on peoples' own behaviors. Taken together, our findings suggest that the PGI of depression largely picks up the risk of behaviorally-influenced adversities, but to a lesser degree also captures other environmental influences. The results invite further exploration into the behavioral and interactional processes that lie along the pathways intervening between genetic differences and wellbeing.
Collapse
Affiliation(s)
- Tamkinat Rauf
- Department of Sociology, University of Wisconsin-Madison, USA.
| | | |
Collapse
|
48
|
Perez-Carpena P, Lopez-Escamez JA, Gallego-Martinez Á. A Systematic Review on the Genetic Contribution to Tinnitus. J Assoc Res Otolaryngol 2024; 25:13-33. [PMID: 38334885 PMCID: PMC10907330 DOI: 10.1007/s10162-024-00925-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/31/2023] [Indexed: 02/10/2024] Open
Abstract
PURPOSE To assess the available evidence to support a genetic contribution and define the role of common and rare variants in tinnitus. METHODS After a systematic search and quality assessment, 31 records including 383,063 patients were selected (14 epidemiological studies and 17 genetic association studies). General information on the sample size, age, sex, tinnitus prevalence, severe tinnitus distribution, and sensorineural hearing loss was retrieved. Studies that did not include data on hearing assessment were excluded. Relative frequencies were used for qualitative variables to compare different studies and to obtain average values. Genetic variants and genes were listed and clustered according to their potential role in tinnitus development. RESULTS The average prevalence of tinnitus estimated from population-based studies was 26.3% for any tinnitus, and 20% of patients with tinnitus reported it as an annoying symptom. One study has reported population-specific differences in the prevalence of tinnitus, the white ancestry being the population with a higher prevalence. Genome-wide association studies have identified and replicated two common variants in the Chinese population (rs2846071; rs4149577) in the intron of TNFRSF1A, associated with noise-induced tinnitus. Moreover, gene burden analyses in sequencing data from Spanish and Swede patients with severe tinnitus have identified and replicated ANK2, AKAP9, and TSC2 genes. CONCLUSIONS The genetic contribution to tinnitus is starting to be revealed and it shows population-specific effects in European and Asian populations. The common allelic variants associated with tinnitus that showed replication are associated with noise-induced tinnitus. Although severe tinnitus has been associated with rare variants with large effect, their role on hearing or hyperacusis has not been established.
Collapse
Affiliation(s)
- Patricia Perez-Carpena
- Otology and Neurotology Group CTS495, Division of Otolaryngology, Department of Surgery, Instituto de Investigación Biosanitaria, Ibs.GRANADA, Universidad de Granada, Granada, Spain.
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain.
- Department of Otolaryngology, Instituto de Investigación Biosanitaria Ibs.GRANADA, Hospital Universitario Virgen de Las Nieves, Granada, Spain.
| | - Jose A Lopez-Escamez
- Otology and Neurotology Group CTS495, Division of Otolaryngology, Department of Surgery, Instituto de Investigación Biosanitaria, Ibs.GRANADA, Universidad de Granada, Granada, Spain.
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain.
- Meniere's Disease Neuroscience Research Program, Faculty of Medicine & Health, School of Medical Sciences, The Kolling Institute, University of Sydney, Sydney, NSW, Australia.
| | - Álvaro Gallego-Martinez
- Otology and Neurotology Group CTS495, Division of Otolaryngology, Department of Surgery, Instituto de Investigación Biosanitaria, Ibs.GRANADA, Universidad de Granada, Granada, Spain
- Sensorineural Pathology Programme, Centro de Investigación Biomédica en Red en Enfermedades Raras, CIBERER, Madrid, Spain
| |
Collapse
|
49
|
Kimmins S, Anderson RA, Barratt CLR, Behre HM, Catford SR, De Jonge CJ, Delbes G, Eisenberg ML, Garrido N, Houston BJ, Jørgensen N, Krausz C, Lismer A, McLachlan RI, Minhas S, Moss T, Pacey A, Priskorn L, Schlatt S, Trasler J, Trasande L, Tüttelmann F, Vazquez-Levin MH, Veltman JA, Zhang F, O'Bryan MK. Frequency, morbidity and equity - the case for increased research on male fertility. Nat Rev Urol 2024; 21:102-124. [PMID: 37828407 DOI: 10.1038/s41585-023-00820-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2023] [Indexed: 10/14/2023]
Abstract
Currently, most men with infertility cannot be given an aetiology, which reflects a lack of knowledge around gamete production and how it is affected by genetics and the environment. A failure to recognize the burden of male infertility and its potential as a biomarker for systemic illness exists. The absence of such knowledge results in patients generally being treated as a uniform group, for whom the strategy is to bypass the causality using medically assisted reproduction (MAR) techniques. In doing so, opportunities to prevent co-morbidity are missed and the burden of MAR is shifted to the woman. To advance understanding of men's reproductive health, longitudinal and multi-national centres for data and sample collection are essential. Such programmes must enable an integrated view of the consequences of genetics, epigenetics and environmental factors on fertility and offspring health. Definition and possible amelioration of the consequences of MAR for conceived children are needed. Inherent in this statement is the necessity to promote fertility restoration and/or use the least invasive MAR strategy available. To achieve this aim, protocols must be rigorously tested and the move towards personalized medicine encouraged. Equally, education of the public, governments and clinicians on the frequency and consequences of infertility is needed. Health options, including male contraceptives, must be expanded, and the opportunities encompassed in such investment understood. The pressing questions related to male reproductive health, spanning the spectrum of andrology are identified in the Expert Recommendation.
Collapse
Affiliation(s)
- Sarah Kimmins
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
- The Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
- The Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, Quebec, Canada
| | - Richard A Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - Christopher L R Barratt
- Division of Systems Medicine, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Hermann M Behre
- Center for Reproductive Medicine and Andrology, University Hospital, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Sarah R Catford
- Hudson Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Obstetrics and Gynaecology, The Royal Women's Hospital, Melbourne, Victoria, Australia
| | | | - Geraldine Delbes
- Institut National de la Recherche Scientifique, Centre Armand-Frappier Sante Biotechnologie, Laval, Quebec, Canada
| | - Michael L Eisenberg
- Department of Urology and Obstetrics and Gynecology, Stanford University, Stanford, CA, USA
| | - Nicolas Garrido
- IVI Foundation, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Brendan J Houston
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Melbourne, Australia
| | - Niels Jørgensen
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Csilla Krausz
- Department of Experimental and Clinical Biomedical Sciences, 'Mario Serio', University of Florence, University Hospital of Careggi Florence, Florence, Italy
| | - Ariane Lismer
- Department of Pharmacology and Therapeutics, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Robert I McLachlan
- Hudson Institute of Medical Research and the Department of Obstetrics and Gynaecology, Monash University, Melbourne, Australia
- Monash IVF Group, Richmond, Victoria, Australia
| | - Suks Minhas
- Department of Surgery and Cancer Imperial, London, UK
| | - Tim Moss
- Healthy Male and the Department of Obstetrics and Gynaecology, Monash University, Melbourne, Victoria, Australia
| | - Allan Pacey
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Lærke Priskorn
- Department of Growth and Reproduction, International Center for Research and Research Training in Endocrine Disruption of Male Reproduction and Child Health, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Stefan Schlatt
- Centre for Reproductive Medicine and Andrology, University of Münster, Münster, Germany
| | - Jacquetta Trasler
- Departments of Paediatrics, Human Genetics and Pharmacology & Therapeutics, McGill University and Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Leonardo Trasande
- Center for the Investigation of Environmental Hazards, Department of Paediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Frank Tüttelmann
- Institute of Reproductive Genetics, University of Münster, Münster, Germany
| | - Mónica Hebe Vazquez-Levin
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas de Argentina, Fundación IBYME, Buenos Aires, Argentina
| | - Joris A Veltman
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Feng Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Moira K O'Bryan
- School of BioSciences and Bio21 Institute, The University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
50
|
Gunter NB, Gebre RK, Graff-Radford J, Heckman MG, Jack CR, Lowe VJ, Knopman DS, Petersen RC, Ross OA, Vemuri P, Ramanan VK. Machine Learning Models of Polygenic Risk for Enhanced Prediction of Alzheimer Disease Endophenotypes. Neurol Genet 2024; 10:e200120. [PMID: 38250184 PMCID: PMC10798228 DOI: 10.1212/nxg.0000000000200120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/01/2023] [Indexed: 01/23/2024]
Abstract
Background and Objectives Alzheimer disease (AD) has a polygenic architecture, for which genome-wide association studies (GWAS) have helped elucidate sequence variants (SVs) influencing susceptibility. Polygenic risk score (PRS) approaches show promise for generating summary measures of inherited risk for clinical AD based on the effects of APOE and other GWAS hits. However, existing PRS approaches, based on traditional regression models, explain only modest variation in AD dementia risk and AD-related endophenotypes. We hypothesized that machine learning (ML) models of polygenic risk (ML-PRS) could outperform standard regression-based PRS methods and therefore have the potential for greater clinical utility. Methods We analyzed combined data from the Mayo Clinic Study of Aging (n = 1,791) and the Alzheimer's Disease Neuroimaging Initiative (n = 864). An AD PRS was computed for each participant using the top common SVs obtained from a large AD dementia GWAS. In parallel, ML models were trained using those SV genotypes, with amyloid PET burden as the primary outcome. Secondary outcomes included amyloid PET positivity and clinical diagnosis (cognitively unimpaired vs impaired). We compared performance between ML-PRS and standard PRS across 100 training sessions with different data splits. In each session, data were split into 80% training and 20% testing, and then five-fold cross-validation was used within the training set to ensure the best model was produced for testing. We also applied permutation importance techniques to assess which genetic factors contributed most to outcome prediction. Results ML-PRS models outperformed the AD PRS (r2 = 0.28 vs r2 = 0.24 in test set) in explaining variation in amyloid PET burden. Among ML approaches, methods accounting for nonlinear genetic influences were superior to linear methods. ML-PRS models were also more accurate when predicting amyloid PET positivity (area under the curve [AUC] = 0.80 vs AUC = 0.63) and the presence of cognitive impairment (AUC = 0.75 vs AUC = 0.54) compared with the standard PRS. Discussion We found that ML-PRS approaches improved upon standard PRS for prediction of AD endophenotypes, partly related to improved accounting for nonlinear effects of genetic susceptibility alleles. Further adaptations of the ML-PRS framework could help to close the gap of remaining unexplained heritability for AD and therefore facilitate more accurate presymptomatic and early-stage risk stratification for clinical decision-making.
Collapse
Affiliation(s)
- Nathaniel B Gunter
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Robel K Gebre
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Jonathan Graff-Radford
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Michael G Heckman
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Clifford R Jack
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Val J Lowe
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - David S Knopman
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Ronald C Petersen
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Owen A Ross
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Prashanthi Vemuri
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| | - Vijay K Ramanan
- From the Departments of Radiology (N.B.G., R.K.G., C.R.J., V.J.L., P.V.), Neurology (J.G.-R., D.S.K., R.C.P., V.K.R.), and Quantitative Health Sciences (R.C.P.), Mayo Clinic Rochester, MN; and Departments of Quantitative Health Sciences (M.G.H.), Neuroscience (O.A.R.), and Clinical Genomics (O.A.R.), Mayo Clinic Florida, Jacksonville
| |
Collapse
|