1
|
Halvorsen MW, de Schipper E, Bäckman J, Strom NI, Hagen K, Lindblad-Toh K, Karlsson EK, Pedersen NL, Wallert J, Bulik CM, Fundín B, Landén M, Kvale G, Hansen B, Haavik J, Mattheisen M, Rück C, Mataix-Cols D, Crowley JJ. A burden of rare copy number variants in obsessive-compulsive disorder. Mol Psychiatry 2024:10.1038/s41380-024-02763-7. [PMID: 39463448 DOI: 10.1038/s41380-024-02763-7] [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: 12/13/2023] [Revised: 08/23/2024] [Accepted: 09/23/2024] [Indexed: 10/29/2024]
Abstract
Current genetic research on obsessive-compulsive disorder (OCD) supports contributions to risk specifically from common single nucleotide variants (SNVs), along with rare coding SNVs and small insertion-deletions (indels). The contribution to OCD risk from rare copy number variants (CNVs), however, has not been formally assessed at a similar scale. Here we describe an analysis of rare CNVs called from genotype array data in 2248 deeply phenotyped OCD cases and 3608 unaffected controls from Sweden and Norway. Cases carry an elevated burden of CNVs ≥30 kb in size (OR = 1.12, P = 1.77 × 10-3). The excess rate of these CNVs in cases versus controls was around 0.07 (95% CI 0.02-0.11, P = 2.58 × 10-3). This signal was largely driven by CNVs overlapping protein-coding regions (OR = 1.19, P = 3.08 × 10-4), particularly deletions impacting loss-of-function intolerant genes (pLI >0.995, OR = 4.12, P = 2.54 × 10-5). We did not identify any specific locus where CNV burden was associated with OCD case status at genome-wide significance, but we noted non-random recurrence of CNV deletions in cases (permutation P = 2.60 × 10-3). In cases where sufficient clinical data were available (n = 1612) we found that carriers of neurodevelopmental duplications were more likely to have comorbid autism (P < 0.001), and that carriers of deletions overlapping neurodevelopmental genes had lower treatment response (P = 0.02). The results demonstrate a contribution of rare CNVs to OCD risk, and suggest that studies of rare coding variation in OCD would have increased power to identify risk genes if this class of variation were incorporated into formal tests.
Collapse
Affiliation(s)
- Matthew W Halvorsen
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Sweden.
| | - Elles de Schipper
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Sweden
| | - Julia Bäckman
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Sweden
| | - Nora I Strom
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Sweden
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Kristen Hagen
- Department of Psychiatry, Molde Hospital, Molde, Norway
- Department of Mental Health, Norwegian University of Science and Technology, Trondheim, Norway
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Kerstin Lindblad-Toh
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, 751 32, Uppsala, Sweden
- Broad Institute of MIT and Harvard, Cambridge, MA, 02139, USA
| | - Elinor K Karlsson
- Broad Institute of MIT and Harvard, Cambridge, MA, 02139, USA
- Program in Bioinformatics and Integrative Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Nancy L Pedersen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - John Wallert
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Sweden
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bengt Fundín
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Mikael Landén
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden
| | - Gerd Kvale
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Psychology, University of Bergen, Bergen, Norway
| | - Bjarne Hansen
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Center for Crisis Psychology, Faculty of Psychology, University of Bergen, Bergen, Norway
| | - Jan Haavik
- Bergen Center for Brain Plasticity, Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Manuel Mattheisen
- Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich, Germany
- Dalhousie University, Department of Community Health and Epidemiology & Faculty of Computer Science, Halifax, Nova Scotia, Canada
| | - Christian Rück
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Sweden
| | - David Mataix-Cols
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Sweden
- Department of Clinical Sciences, Lund University, Lund, Sweden
| | - James J Crowley
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, Region Stockholm, Sweden
| |
Collapse
|
2
|
Harris L, McDonagh EM, Zhang X, Fawcett K, Foreman A, Daneck P, Sergouniotis PI, Parkinson H, Mazzarotto F, Inouye M, Hollox EJ, Birney E, Fitzgerald T. Genome-wide association testing beyond SNPs. Nat Rev Genet 2024:10.1038/s41576-024-00778-y. [PMID: 39375560 DOI: 10.1038/s41576-024-00778-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2024] [Indexed: 10/09/2024]
Abstract
Decades of genetic association testing in human cohorts have provided important insights into the genetic architecture and biological underpinnings of complex traits and diseases. However, for certain traits, genome-wide association studies (GWAS) for common SNPs are approaching signal saturation, which underscores the need to explore other types of genetic variation to understand the genetic basis of traits and diseases. Copy number variation (CNV) is an important source of heritability that is well known to functionally affect human traits. Recent technological and computational advances enable the large-scale, genome-wide evaluation of CNVs, with implications for downstream applications such as polygenic risk scoring and drug target identification. Here, we review the current state of CNV-GWAS, discuss current limitations in resource infrastructure that need to be overcome to enable the wider uptake of CNV-GWAS results, highlight emerging opportunities and suggest guidelines and standards for future GWAS for genetic variation beyond SNPs at scale.
Collapse
Affiliation(s)
- Laura Harris
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Ellen M McDonagh
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Xiaolei Zhang
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Katherine Fawcett
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
- Department of Population Health Sciences, University of Leicester, Leicester, UK
| | - Amy Foreman
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Petr Daneck
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Panagiotis I Sergouniotis
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, University of Manchester, Manchester, UK
| | - Helen Parkinson
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Francesco Mazzarotto
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Michael Inouye
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Cambridge Baker Systems Genomics Initiative, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Edward J Hollox
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Ewan Birney
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK
| | - Tomas Fitzgerald
- European Molecular Biology Laboratory (EMBL), European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, UK.
| |
Collapse
|
3
|
Minnai F, Noci S, Esposito M, Schneider MA, Kobinger S, Eichhorn M, Winter H, Hoffmann H, Kriegsmann M, Incarbone MA, Mattioni G, Tosi D, Muley T, Dragani TA, Colombo F. Germline Polymorphisms Associated with Overall Survival in Lung Adenocarcinoma: Genome-Wide Analysis. Cancers (Basel) 2024; 16:3264. [PMID: 39409885 PMCID: PMC11475969 DOI: 10.3390/cancers16193264] [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: 08/19/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND/OBJECTIVES Lung cancer remains a global health concern, with substantial variation in patient survival. Despite advances in detection and treatment, the genetic basis for the divergent outcomes is not understood. We investigated germline polymorphisms that modulate overall survival in 1464 surgically resected lung adenocarcinoma patients. METHODS A multivariable Cox proportional hazard model was used to assess the association of more than seven million polymorphisms with overall survival at the 60-month follow-up, considering age, sex, pathological stage, decade of surgery and principal components as covariates. Genes in which variants were identified were studied in silico to investigate functional roles. RESULTS Six germline variants passed the genome-wide significance threshold. These single nucleotide polymorphisms were mapped to non-coding (intronic) regions on chromosomes 2, 3, and 5. The minor alleles of rs13000315, rs151212827, and rs190923216 (chr. 2, 3 and 5, respectively) were found to be independent negative prognostic factors. All six variants have been reported to regulate the expression of nine genes, seven of which are protein-coding, in different tissues. Survival-associated variants on chromosomes 2 and 3 were already reported to regulate the expression of NT5DC2 and NAGK, with high expression associated with the minor alleles. High NT5DC2 and NAGK expression in lung adenocarcinoma tissue was already shown to correlate with poor overall survival. CONCLUSIONS This study highlights a potential regulatory role of the identified polymorphisms in influencing outcome and suggests a mechanistic link between these variants, gene expression regulation, and lung adenocarcinoma prognosis. Validation and functional studies are warranted to elucidate the mechanisms underlying these associations.
Collapse
Affiliation(s)
- Francesca Minnai
- Institute for Biomedical Technologies, National Research Council, Segrate, 20054 Milan, Italy (F.C.)
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, 20122 Milan, Italy
| | - Sara Noci
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Martina Esposito
- Institute for Biomedical Technologies, National Research Council, Segrate, 20054 Milan, Italy (F.C.)
| | - Marc A. Schneider
- Translational Research Unit (STF), Thoraxklinik, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Sonja Kobinger
- Department of Thoracic Surgery, Thoraxklinik, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Martin Eichhorn
- Department of Thoracic Surgery, Thoraxklinik, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Department of Thoracic Surgery, Thoraxklinik, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Hans Hoffmann
- Department of Thoracic Surgery, Klinikum Rechts der Isar, Technische Universität München, 80333 Munich, Germany
| | - Mark Kriegsmann
- Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
- Institute of Pathology, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Matteo A. Incarbone
- Department of Surgery, Ospedale San Giuseppe, IRCCS Multimedica, 20099 Milan, Italy
| | - Giovanni Mattioni
- Thoracic Surgery and Lung Transplantation Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Davide Tosi
- Thoracic Surgery and Lung Transplantation Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Thomas Muley
- Translational Research Unit (STF), Thoraxklinik, University Hospital Heidelberg, 69120 Heidelberg, Germany
- Translational Lung Research Center (TLRC), German Center for Lung Research (DZL), 69120 Heidelberg, Germany
| | - Tommaso A. Dragani
- Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy
| | - Francesca Colombo
- Institute for Biomedical Technologies, National Research Council, Segrate, 20054 Milan, Italy (F.C.)
| |
Collapse
|
4
|
Lobato-Martinez E, Muriel-Serrano J, García-Payá E, Gonzalez-de-la-Aleja P, Garcia-Sevila R, Navarro-de-Miguel M, Marco-de-la-Calle F, Ramos-Rincon JM, Sanchez-Martinez R. Association of Human Leukocyte Antigen Alleles with COVID-19 Severity and Mortality in a Spanish Population. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1392. [PMID: 39336433 PMCID: PMC11434301 DOI: 10.3390/medicina60091392] [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] [Received: 07/04/2024] [Revised: 08/12/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024]
Abstract
Background and Objectives: The aim of the following cross-sectional study is to determine the association between human leukocyte antigen (HLA) alleles and outcomes in patients presenting to the emergency department (ED) with SARS-CoV-2 infection. Methods and Materials: Genotyping was made using the Axiom Human Genotyping SARS-CoV-2 Research Array. Statistical analysis was made with Fisher's exact test and multivariable logistic regression, adjusted for sex, age and clinical variables. Results: Of 190 patients, 11.1% were discharged from the ED; 57.9% were admitted to the COVID-19 ward, without intensive care unit (ICU) admission; 15.3% survived an ICU admission; and 15.8% died. After multivariable analysis, two HLA alleles protected against hospital admission (HLA-C*05:01, adjusted odds ratio [aOR] 0.2, 95% confidence interval [CI] 0.055-0.731; and HLA-DQB1*02:02, aOR 0.046, CI 0.002-0.871) and one was associated with higher risk for ICU admission or death (HLA-DQA1*05:01, aOR 2.517, CI 1.086-5.833). Conclusions: In this population, HLA-C*05:01 and HLA-DQB1*02:02 are associated with a protective effect against hospital admission and HLA-DQA1*05:01 is associated with higher risk of ICU admission or death in the multivariable analysis. This may help stratify risk in COVID-19 patients.
Collapse
Affiliation(s)
- Ester Lobato-Martinez
- Internal Medicine Department, Dr. Balmis Universitary General Hospital, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Centro de Diagnóstico, Edif Gris, Planta 5ª, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
| | - Javier Muriel-Serrano
- Alicante Institute for Health and Biomedical Research (ISABIAL), Centro de Diagnóstico, Edif Gris, Planta 5ª, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
| | - Elena García-Payá
- Alicante Institute for Health and Biomedical Research (ISABIAL), Centro de Diagnóstico, Edif Gris, Planta 5ª, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Analysis Department, Dr. Balmis Universitary General Hospital, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
| | - Pilar Gonzalez-de-la-Aleja
- Alicante Institute for Health and Biomedical Research (ISABIAL), Centro de Diagnóstico, Edif Gris, Planta 5ª, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Infectious Diseases Unit, Dr. Balmis Universitary General Hospital, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
| | - Raquel Garcia-Sevila
- Alicante Institute for Health and Biomedical Research (ISABIAL), Centro de Diagnóstico, Edif Gris, Planta 5ª, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Pneumology Department, Dr. Balmis Universitary General Hospital, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
| | - Mercedes Navarro-de-Miguel
- Alicante Institute for Health and Biomedical Research (ISABIAL), Centro de Diagnóstico, Edif Gris, Planta 5ª, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Analysis Department, Dr. Balmis Universitary General Hospital, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
| | - Francisco Marco-de-la-Calle
- Alicante Institute for Health and Biomedical Research (ISABIAL), Centro de Diagnóstico, Edif Gris, Planta 5ª, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Immunology Department, Dr. Balmis Universitary General Hospital, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
| | - Jose-Manuel Ramos-Rincon
- Internal Medicine Department, Dr. Balmis Universitary General Hospital, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Centro de Diagnóstico, Edif Gris, Planta 5ª, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Medicine Department, Miguel Hernández University, N-332, 87, 03550 Alicante, Spain
| | - Rosario Sanchez-Martinez
- Internal Medicine Department, Dr. Balmis Universitary General Hospital, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Centro de Diagnóstico, Edif Gris, Planta 5ª, Avenida Pintor Baeza, 12, 03010 Alicante, Spain
- Clinical Medicine Department, Miguel Hernández University, N-332, 87, 03550 Alicante, Spain
| |
Collapse
|
5
|
Delabays B, Trajanoska K, Walonoski J, Mooser V. Cardiovascular Pharmacogenetics: From Discovery of Genetic Association to Clinical Adoption of Derived Test. Pharmacol Rev 2024; 76:791-827. [PMID: 39122647 DOI: 10.1124/pharmrev.123.000750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 04/24/2024] [Accepted: 05/28/2024] [Indexed: 08/12/2024] Open
Abstract
Recent breakthroughs in human genetics and in information technologies have markedly expanded our understanding at the molecular level of the response to drugs, i.e., pharmacogenetics (PGx), across therapy areas. This review is restricted to PGx for cardiovascular (CV) drugs. First, we examined the PGx information in the labels approved by regulatory agencies in Europe, Japan, and North America and related recommendations from expert panels. Out of 221 marketed CV drugs, 36 had PGx information in their labels approved by one or more agencies. The level of annotations and recommendations varied markedly between agencies and expert panels. Clopidogrel is the only CV drug with consistent PGx recommendation (i.e., "actionable"). This situation prompted us to dissect the steps from discovery of a PGx association to clinical translation. We found 101 genome-wide association studies that investigated the response to CV drugs or drug classes. These studies reported significant associations for 48 PGx traits mapping to 306 genes. Six of these 306 genes are mentioned in the corresponding PGx labels or recommendations for CV drugs. Genomic analyses also highlighted the wide between-population differences in risk allele frequencies and the individual load of actionable PGx variants. Given the high attrition rate and the long road to clinical translation, additional work is warranted to identify and validate PGx variants for more CV drugs across diverse populations and to demonstrate the utility of PGx testing. To that end, pre-emptive PGx combining genomic profiling with electronic medical records opens unprecedented opportunities to improve healthcare, for CV diseases and beyond. SIGNIFICANCE STATEMENT: Despite spectacular breakthroughs in human molecular genetics and information technologies, consistent evidence supporting PGx testing in the cardiovascular area is limited to a few drugs. Additional work is warranted to discover and validate new PGx markers and demonstrate their utility. Pre-emptive PGx combining genomic profiling with electronic medical records opens unprecedented opportunities to improve healthcare, for CV diseases and beyond.
Collapse
Affiliation(s)
- Benoît Delabays
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada (B.D., K.T., V.M.); and Medeloop Inc., Palo Alto, California, and Montreal, QC, Canada (J.W.)
| | - Katerina Trajanoska
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada (B.D., K.T., V.M.); and Medeloop Inc., Palo Alto, California, and Montreal, QC, Canada (J.W.)
| | - Joshua Walonoski
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada (B.D., K.T., V.M.); and Medeloop Inc., Palo Alto, California, and Montreal, QC, Canada (J.W.)
| | - Vincent Mooser
- Canada Excellence Research Chair in Genomic Medicine, Victor Phillip Dahdaleh Institute of Genomic Medicine, Department of Human Genetics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QC, Canada (B.D., K.T., V.M.); and Medeloop Inc., Palo Alto, California, and Montreal, QC, Canada (J.W.)
| |
Collapse
|
6
|
Stikker B, Trap L, Sedaghati-Khayat B, de Bruijn MJW, van Ijcken WFJ, de Roos E, Ikram A, Hendriks RW, Brusselle G, van Rooij J, Stadhouders R. Epigenomic partitioning of a polygenic risk score for asthma reveals distinct genetically driven disease pathways. Eur Respir J 2024; 64:2302059. [PMID: 38901884 PMCID: PMC11358516 DOI: 10.1183/13993003.02059-2023] [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: 11/22/2023] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Individual differences in susceptibility to developing asthma, a heterogeneous chronic inflammatory lung disease, are poorly understood. Whether genetics can predict asthma risk and how genetic variants modulate the complex pathophysiology of asthma are still debated. AIM To build polygenic risk scores for asthma risk prediction and epigenomically link predictive genetic variants to pathophysiological mechanisms. METHODS Restricted polygenic risk scores were constructed using single nucleotide variants derived from genome-wide association studies and validated using data generated in the Rotterdam Study, a Dutch prospective cohort of 14 926 individuals. Outcomes used were asthma, childhood-onset asthma, adulthood-onset asthma, eosinophilic asthma and asthma exacerbations. Genome-wide chromatin analysis data from 19 disease-relevant cell types were used for epigenomic polygenic risk score partitioning. RESULTS The polygenic risk scores obtained predicted asthma and related outcomes, with the strongest associations observed for childhood-onset asthma (2.55 odds ratios per polygenic risk score standard deviation, area under the curve of 0.760). Polygenic risk scores allowed for the classification of individuals into high-risk and low-risk groups. Polygenic risk score partitioning using epigenomic profiles identified five clusters of variants within putative gene regulatory regions linked to specific asthma-relevant cells, genes and biological pathways. CONCLUSIONS Polygenic risk scores were associated with asthma(-related traits) in a Dutch prospective cohort, with substantially higher predictive power observed for childhood-onset than adult-onset asthma. Importantly, polygenic risk score variants could be epigenomically partitioned into clusters of regulatory variants with different pathophysiological association patterns and effect estimates, which likely represent distinct genetically driven disease pathways. Our findings have potential implications for personalised risk mitigation and treatment strategies.
Collapse
Affiliation(s)
- Bernard Stikker
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Lianne Trap
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- L. Trap and B. Sedaghati-Khayat made an equal contribution to this study
| | - Bahar Sedaghati-Khayat
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- L. Trap and B. Sedaghati-Khayat made an equal contribution to this study
| | - Marjolein J W de Bruijn
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Wilfred F J van Ijcken
- Center for Biomics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Emmely de Roos
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Arfan Ikram
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Guy Brusselle
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jeroen van Rooij
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- J. van Rooij and R. Stadhouders contributed equally to this article as lead authors and supervised the work
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- J. van Rooij and R. Stadhouders contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
7
|
Stiehler S, Sembill S, Schleicher O, Marx M, Rauh M, Krumbholz M, Karow A, Suttorp M, Woelfle J, Maj C, Metzler M. Imatinib treatment and longitudinal growth in pediatric patients with chronic myeloid leukemia: influence of demographic, pharmacological, and genetic factors in the German CML-PAED cohort. Haematologica 2024; 109:2555-2563. [PMID: 38497150 PMCID: PMC11290534 DOI: 10.3324/haematol.2023.284668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/05/2024] [Indexed: 03/19/2024] Open
Abstract
In children and adolescents, impaired growth due to tyrosine kinase inhibitor therapy remains an insufficiently studied adverse effect. This study examines demographic, pharmacological, and genetic factors associated with impaired longitudinal growth in a uniform pediatric cohort treated with imatinib. We analyzed 94 pediatric patients with chronic myeloid leukemia (CML) diagnosed in the chronic phase and treated with imatinib for >12 months who participated in the Germany-wide CML-PAEDII study between February 2006 and February 2021 (clinicaltrials gov. Identifier: NCT00445822). During imatinib treatment, significant height reduction occurred, with medians of -0.35 standard deviation score (SDS) at 12 months and -0.76 SDS at 24 months. Cumulative height SDS change (Δ height SDS) showed a more pronounced effect in prepubertal patients during the first year but were similar between prepubertal and pubertal subgroups by the second year (-0.55 vs. -0.50). From months 12 to 18 on imatinib, only 18% patients achieved individually longitudinal growth adequate to the growth standard (Δ height SDS ≥0). When patients were divided into two subgroups based on median Δ height SDS (classifier Δ height SDS > or ≤-0.37) after 1 year on imatinib therapy, cohort 1 (Δ height SDS ≤-0.37) showed younger age at diagnosis, a higher proportion of prepubertal children, but also better treatment response and higher imatinib serum levels. Exploring the association of growth parameters with pharmacokinetically relevant single nucleotide polymorphisms, known for affecting imatinib response, showed no correlation. This retrospective study provides new insights into imatinib-related growth impairment. We emphasize the importance of optimizing treatment strategies for pediatric patients to realize their maximum growth potential.
Collapse
Affiliation(s)
- Sophie Stiehler
- Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg
| | - Stephanie Sembill
- Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen.
| | - Oliver Schleicher
- Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Germany; Department of Gynecology and Obstetrics, University Hospital Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg
| | - Michaela Marx
- Pediatric Endocrinology, Department of Pediatrics and Adolescent Medicine, University Children's Hospital, Friedrich-Alexander-Universität, Erlangen-Nürnberg
| | - Manfred Rauh
- Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg
| | - Manuela Krumbholz
- Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen
| | - Axel Karow
- Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen
| | - Meinolf Suttorp
- Pediatric Hemato-Oncology, Medical Faculty, Technical University Dresden, Dresden
| | - Joachim Woelfle
- Pediatric Endocrinology, Department of Pediatrics and Adolescent Medicine, University Children's Hospital, Friedrich-Alexander-Universität, Erlangen-Nürnberg
| | - Carlo Maj
- Centre for Human Genetics, University of Marburg, Marburg
| | - Markus Metzler
- Pediatric Oncology and Hematology, Department of Pediatrics and Adolescent Medicine, University Hospital Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg, Germany; Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen
| |
Collapse
|
8
|
Martinez KL, Klein A, Martin JR, Sampson CU, Giles JB, Beck ML, Bhakta K, Quatraro G, Farol J, Karnes JH. Disparities in ABO blood type determination across diverse ancestries: a systematic review and validation in the All of Us Research Program. J Am Med Inform Assoc 2024:ocae161. [PMID: 38917427 DOI: 10.1093/jamia/ocae161] [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: 03/20/2024] [Revised: 05/02/2024] [Accepted: 06/19/2024] [Indexed: 06/27/2024] Open
Abstract
OBJECTIVES ABO blood types have widespread clinical use and robust associations with disease. The purpose of this study is to evaluate the portability and suitability of tag single-nucleotide polymorphisms (tSNPs) used to determine ABO alleles and blood types across diverse populations in published literature. MATERIALS AND METHODS Bibliographic databases were searched for studies using tSNPs to determine ABO alleles. We calculated linkage between tSNPs and functional variants across inferred continental ancestry groups from 1000 Genomes. We compared r2 across ancestry and assessed real-world consequences by comparing tSNP-derived blood types to serology in a diverse population from the All of Us Research Program. RESULTS Linkage between functional variants and O allele tSNPs was significantly lower in African (median r2 = 0.443) compared to East Asian (r2 = 0.946, P = 1.1 × 10-5) and European (r2 = 0.869, P = .023) populations. In All of Us, discordance between tSNP-derived blood types and serology was high across all SNPs in African ancestry individuals and linkage was strongly correlated with discordance across all ancestries (ρ = -0.90, P = 3.08 × 10-23). DISCUSSION Many studies determine ABO blood types using tSNPs. However, tSNPs with low linkage disequilibrium promote misinference of ABO blood types, particularly in diverse populations. We observe common use of inappropriate tSNPs to determine ABO blood type, particularly for O alleles and with some tSNPs mistyping up to 58% of individuals. CONCLUSION Our results highlight the lack of transferability of tSNPs across ancestries and potential exacerbation of disparities in genomic research for underrepresented populations. This is especially relevant as more diverse cohorts are made publicly available.
Collapse
Affiliation(s)
- Kiana L Martinez
- Department of Pharmacy Practice and Science, The University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ 85721, United States
| | - Andrew Klein
- Department of Pharmacy Practice and Science, The University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ 85721, United States
| | - Jennifer R Martin
- Department of Pharmacy Practice and Science, The University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ 85721, United States
- Department of the University of Arizona Health Sciences Library, The University of Arizona, Tucson, AZ 85721, United States
| | - Chinwuwanuju U Sampson
- Department of Pharmacy Practice and Science, The University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ 85721, United States
| | - Jason B Giles
- Department of Pharmacy Practice and Science, The University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ 85721, United States
| | - Madison L Beck
- Department of Pharmacy Practice and Science, The University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ 85721, United States
| | - Krupa Bhakta
- Department of Pharmacy Practice and Science, The University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ 85721, United States
| | - Gino Quatraro
- Department of Pharmacy Practice and Science, The University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ 85721, United States
| | - Juvie Farol
- Department of Clinical and Translational Science, The University of Arizona College of Medicine, Tucson, AZ 85721, United States
| | - Jason H Karnes
- Department of Pharmacy Practice and Science, The University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ 85721, United States
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| |
Collapse
|
9
|
Santos R, Moreno-Torres V, Pintos I, Corral O, de Mendoza C, Soriano V, Corpas M. Low-coverage whole genome sequencing for a highly selective cohort of severe COVID-19 patients. GIGABYTE 2024; 2024:gigabyte127. [PMID: 38948510 PMCID: PMC11211761 DOI: 10.46471/gigabyte.127] [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: 09/25/2023] [Accepted: 06/04/2024] [Indexed: 07/02/2024] Open
Abstract
Despite the advances in genetic marker identification associated with severe COVID-19, the full genetic characterisation of the disease remains elusive. This study explores imputation in low-coverage whole genome sequencing for a severe COVID-19 patient cohort. We generated a dataset of 79 imputed variant call format files using the GLIMPSE1 tool, each containing an average of 9.5 million single nucleotide variants. Validation revealed a high imputation accuracy (squared Pearson correlation ≍0.97) across sequencing platforms, showcasing GLIMPSE1's ability to confidently impute variants with minor allele frequencies as low as 2% in individuals with Spanish ancestry. We carried out a comprehensive analysis of the patient cohort, examining hospitalisation and intensive care utilisation, sex and age-based differences, and clinical phenotypes using a standardised set of medical terms developed to characterise severe COVID-19 symptoms. The methods and findings presented here can be leveraged for future genomic projects to gain vital insights into health challenges like COVID-19.
Collapse
Affiliation(s)
- Renato Santos
- National Heart & Lung Institute, Imperial College London, London, UK
| | - Víctor Moreno-Torres
- Puerta de Hierro University Hospital & Research Institute, Majadahonda, Madrid, Spain
| | - Ilduara Pintos
- Puerta de Hierro University Hospital & Research Institute, Majadahonda, Madrid, Spain
| | - Octavio Corral
- Health Sciences School & Medical Centre, Universidad Internacional La Rioja (UNIR), Madrid, Spain
| | - Carmen de Mendoza
- Puerta de Hierro University Hospital & Research Institute, Majadahonda, Madrid, Spain
| | - Vicente Soriano
- Health Sciences School & Medical Centre, Universidad Internacional La Rioja (UNIR), Madrid, Spain
| | - Manuel Corpas
- School of Life Sciences, University of Westminster, London, UK
| |
Collapse
|
10
|
Lenz C, Narang A, Bousman CA. Pharmacogenomic allele coverage of genome-wide genotyping arrays: a comparative analysis. Pharmacogenet Genomics 2024; 34:130-134. [PMID: 38359167 DOI: 10.1097/fpc.0000000000000523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
The use of genome-wide genotyping arrays in pharmacogenomics (PGx) research and clinical implementation applications is increasing but it is unclear which arrays are best suited for these applications. Here, we conduct a comparative coverage analysis of PGx alleles included on genome-wide genotyping arrays, with an emphasis on alleles in genes with PGx-based prescribing guidelines. Genomic manifest files for seven arrays including the Axiom Precision Medicine Diversity Array (PMDA), Axiom PMDA Plus, Axiom PangenomiX, Axiom PangenomiX Plus, Infinium Global Screening Array, Infinium Global Diversity Array (GDA) and Infinium GDA with enhanced PGx (GDA-PGx) Array, were evaluated for coverage of 523 star alleles across 19 pharmacogenes included in prescribing guidelines developed by the Clinical Pharmacogenetic Implementation Consortium and Dutch Pharmacogenomics Working Group. Specific attention was given to coverage of the Association of Molecular Pathology's Tier 1 and Tier 2 allele sets for CYP2C9, CYP2C19, CYP2D6, CYP3A4, CYP3A5, NUDT15, TPMT and VKORC1 . Coverage of the examined PGx alleles was highest for the Infinium GDA-PGx (88%), Axiom PangenomiX Plus (77%), Axiom PangenomiX (72%) and Axiom PMDA Plus (70%). Three arrays (Infinium GDA-PGx, Axiom PangenomiX Plus and Axiom PMDA Plus) fully covered the Tier 1 alleles and the Axiom PangenomiX array provided full coverage of Tier 2 alleles. In conclusion, PGx allele coverage varied by gene and array. A superior array for all PGx applications was not identified. Future comparative analyses of genotype data produced by these arrays are needed to determine the robustness of the reported coverage estimates.
Collapse
Affiliation(s)
| | | | - Chad A Bousman
- Alberta Children's Hospital Research Institute
- Department of Medical Genetics
- The Mathison Centre for Mental Health Research and Education, Hotchkiss Brain Institute, Cumming School of Medicine
- Departments of Psychiatry
- Physiology and Pharmacology
- Community Health Sciences, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
11
|
Bose S, Banerjee S, Kumar S, Saha A, Nandy D, Hazra S. Review of applications of artificial intelligence (AI) methods in crop research. J Appl Genet 2024; 65:225-240. [PMID: 38216788 DOI: 10.1007/s13353-023-00826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/23/2023] [Accepted: 12/26/2023] [Indexed: 01/14/2024]
Abstract
Sophisticated and modern crop improvement techniques can bridge the gap for feeding the ever-increasing population. Artificial intelligence (AI) refers to the simulation of human intelligence in machines, which refers to the application of computational algorithms, machine learning (ML) and deep learning (DL) techniques. This is aimed to generalise patterns and relationships from historical data, employing various mathematical optimisation techniques thus making prediction models for facilitating selection of superior genotypes. These techniques are less resource intensive and can solve the problem based on the analysis of large-scale phenotypic datasets. ML for genomic selection (GS) uses high-throughput genotyping technologies to gather genetic information on a large number of markers across the genome. The prediction of GS models is based on the mathematical relation between genotypic and phenotypic data from the training population. ML techniques have emerged as powerful tools for genome editing through analysing large-scale genomic data and facilitating the development of accurate prediction models. Precise phenotyping is a prerequisite to advance crop breeding for solving agricultural production-related issues. ML algorithms can solve this problem through generating predictive models, based on the analysis of large-scale phenotypic datasets. DL models also have the potential reliability of precise phenotyping. This review provides a comprehensive overview on various ML and DL models, their applications, potential to enhance the efficiency, specificity and safety towards advanced crop improvement protocols such as genomic selection, genome editing, along with phenotypic prediction to promote accelerated breeding.
Collapse
Affiliation(s)
- Suvojit Bose
- Department of Vegetables and Spice Crops, Uttar Banga Krishi Viswavidyalaya, Pundibari, Cooch Behar, 736165, West Bengal, India
| | | | - Soumya Kumar
- School of Agricultural Sciences, JIS University, Kolkata, 700109, West Bengal, India
| | - Akash Saha
- School of Agricultural Sciences, JIS University, Kolkata, 700109, West Bengal, India
| | - Debalina Nandy
- School of Agricultural Sciences, JIS University, Kolkata, 700109, West Bengal, India
| | - Soham Hazra
- Department of Agriculture, Brainware University, Barasat, 700125, West Bengal, India.
| |
Collapse
|
12
|
Neuhofer CM, Prokisch H. Digenic Inheritance in Rare Disorders and Mitochondrial Disease-Crossing the Frontier to a More Comprehensive Understanding of Etiology. Int J Mol Sci 2024; 25:4602. [PMID: 38731822 PMCID: PMC11083678 DOI: 10.3390/ijms25094602] [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: 02/13/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 05/13/2024] Open
Abstract
Our understanding of rare disease genetics has been shaped by a monogenic disease model. While the traditional monogenic disease model has been successful in identifying numerous disease-associated genes and significantly enlarged our knowledge in the field of human genetics, it has limitations in explaining phenomena like phenotypic variability and reduced penetrance. Widening the perspective beyond Mendelian inheritance has the potential to enable a better understanding of disease complexity in rare disorders. Digenic inheritance is the simplest instance of a non-Mendelian disorder, characterized by the functional interplay of variants in two disease-contributing genes. Known digenic disease causes show a range of pathomechanisms underlying digenic interplay, including direct and indirect gene product interactions as well as epigenetic modifications. This review aims to systematically explore the background of digenic inheritance in rare disorders, the approaches and challenges when investigating digenic inheritance, and the current evidence for digenic inheritance in mitochondrial disorders.
Collapse
Affiliation(s)
- Christiane M. Neuhofer
- Institute of Human Genetics, University Medical Center, Technical University of Munich, Trogerstr. 32, 81675 Munich, Germany
- Institute of Neurogenomics, Computational Health Center, Helmholtz Centre Munich Neuherberg, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany
- Institute of Human Genetics, Salzburger Landeskliniken, University Hospital of the Paracelsus Medical University, Müllner Hauptstraße 48, 5020 Salzburg, Austria
| | - Holger Prokisch
- Institute of Human Genetics, University Medical Center, Technical University of Munich, Trogerstr. 32, 81675 Munich, Germany
- Institute of Neurogenomics, Computational Health Center, Helmholtz Centre Munich Neuherberg, Ingolstädter Landstraße 1, 85764 Oberschleißheim, Germany
| |
Collapse
|
13
|
Esposito M, Minnai F, Copetti M, Miscio G, Perna R, Piepoli A, De Vincentis G, Benvenuto M, D'Addetta P, Croci S, Baldassarri M, Bruttini M, Fallerini C, Brugnoni R, Cavalcante P, Baggi F, Corsini EMG, Ciusani E, Andreetta F, Dragani TA, Fratelli M, Carella M, Mantegazza RE, Renieri A, Colombo F. Human leukocyte antigen variants associate with BNT162b2 mRNA vaccine response. COMMUNICATIONS MEDICINE 2024; 4:63. [PMID: 38575714 PMCID: PMC10995155 DOI: 10.1038/s43856-024-00490-2] [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: 07/05/2023] [Accepted: 03/21/2024] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Since the beginning of the anti-COVID-19 vaccination campaign, it has become evident that vaccinated subjects exhibit considerable inter-individual variability in the response to the vaccine that could be partly explained by host genetic factors. A recent study reported that the immune response elicited by the Oxford-AstraZeneca vaccine in individuals from the United Kingdom was influenced by a specific allele of the human leukocyte antigen gene HLA-DQB1. METHODS We carried out a genome-wide association study to investigate the genetic determinants of the antibody response to the Pfizer-BioNTech vaccine in an Italian cohort of 1351 subjects recruited in three centers. Linear regressions between normalized antibody levels and genotypes of more than 7 million variants was performed, using sex, age, centers, days between vaccination boost and serological test, and five principal components as covariates. We also analyzed the association between normalized antibody levels and 204 HLA alleles, with the same covariates as above. RESULTS Our study confirms the involvement of the HLA locus and shows significant associations with variants in HLA-A, HLA-DQA1, and HLA-DQB1 genes. In particular, the HLA-A*03:01 allele is the most significantly associated with serum levels of anti-SARS-CoV-2 antibodies. Other alleles, from both major histocompatibility complex class I and II are significantly associated with antibody levels. CONCLUSIONS These results support the hypothesis that HLA genes modulate the response to Pfizer-BioNTech vaccine and highlight the need for genetic studies in diverse populations and for functional studies aimed to elucidate the relationship between HLA-A*03:01 and CD8+ cell response upon Pfizer-BioNTech vaccination.
Collapse
Affiliation(s)
- Martina Esposito
- National Research Council, Institute for Biomedical Technologies, Segrate, MI, Italy
| | - Francesca Minnai
- National Research Council, Institute for Biomedical Technologies, Segrate, MI, Italy
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, Milan, Italy
| | - Massimiliano Copetti
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Giuseppe Miscio
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Rita Perna
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Ada Piepoli
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | | | - Mario Benvenuto
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Paola D'Addetta
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Susanna Croci
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | - Margherita Baldassarri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | - Mirella Bruttini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Chiara Fallerini
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
| | | | | | - Fulvio Baggi
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Emilio Ciusani
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | | | | | - Massimo Carella
- Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | | | - Alessandra Renieri
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Medical Genetics, University of Siena, Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Francesca Colombo
- National Research Council, Institute for Biomedical Technologies, Segrate, MI, Italy.
| |
Collapse
|
14
|
Armoundas AA, Narayan SM, Arnett DK, Spector-Bagdady K, Bennett DA, Celi LA, Friedman PA, Gollob MH, Hall JL, Kwitek AE, Lett E, Menon BK, Sheehan KA, Al-Zaiti SS. Use of Artificial Intelligence in Improving Outcomes in Heart Disease: A Scientific Statement From the American Heart Association. Circulation 2024; 149:e1028-e1050. [PMID: 38415358 PMCID: PMC11042786 DOI: 10.1161/cir.0000000000001201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
A major focus of academia, industry, and global governmental agencies is to develop and apply artificial intelligence and other advanced analytical tools to transform health care delivery. The American Heart Association supports the creation of tools and services that would further the science and practice of precision medicine by enabling more precise approaches to cardiovascular and stroke research, prevention, and care of individuals and populations. Nevertheless, several challenges exist, and few artificial intelligence tools have been shown to improve cardiovascular and stroke care sufficiently to be widely adopted. This scientific statement outlines the current state of the art on the use of artificial intelligence algorithms and data science in the diagnosis, classification, and treatment of cardiovascular disease. It also sets out to advance this mission, focusing on how digital tools and, in particular, artificial intelligence may provide clinical and mechanistic insights, address bias in clinical studies, and facilitate education and implementation science to improve cardiovascular and stroke outcomes. Last, a key objective of this scientific statement is to further the field by identifying best practices, gaps, and challenges for interested stakeholders.
Collapse
|
15
|
Youssef O, Loukola A, Zidi-Mouaffak YHS, Tamlander M, Ruotsalainen S, Kilpeläinen E, Mars N, Ripatti S, Palotie A, Donner K, Carpén O. High-Resolution Genotyping of Formalin-Fixed Tissue Accurately Estimates Polygenic Risk Scores in Human Diseases. J Transl Med 2024; 104:100325. [PMID: 38220043 DOI: 10.1016/j.labinv.2024.100325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 12/11/2023] [Accepted: 01/05/2024] [Indexed: 01/16/2024] Open
Abstract
Formalin-fixed paraffin-embedded (FFPE) tissues stored in biobanks and pathology archives are a vast but underutilized source for molecular studies on different diseases. Beyond being the "gold standard" for preservation of diagnostic human tissues, FFPE samples retain similar genetic information as matching blood samples, which could make FFPE samples an ideal resource for genomic analysis. However, research on this resource has been hindered by the perception that DNA extracted from FFPE samples is of poor quality. Here, we show that germline disease-predisposing variants and polygenic risk scores (PRS) can be identified from FFPE normal tissue (FFPE-NT) DNA with high accuracy. We optimized the performance of FFPE-NT DNA on a genome-wide array containing 657,675 variants. Via a series of testing and validation phases, we established a protocol for FFPE-NT genotyping with results comparable with blood genotyping. The median call rate of FFPE-NT samples in the validation phase was 99.85% (range 98.26%-99.94%) and median concordance with matching blood samples was 99.79% (range 98.85%-99.9%). We also demonstrated that a rare pathogenic PALB2 genetic variant predisposing to cancer can be correctly identified in FFPE-NT samples. We further imputed the FFPE-NT genotype data and calculated the FFPE-NT genome-wide PRS in 3 diseases and 4 disease risk variables. In all cases, FFPE-NT and matching blood PRS were highly concordant (all Pearson's r > 0.95). The ability to precisely genotype FFPE-NT on a genome-wide array enables translational genomics applications of archived FFPE-NT samples with the possibility to link to corresponding phenotypes and longitudinal health data.
Collapse
Affiliation(s)
- Omar Youssef
- Department of Pathology, University of Helsinki, Helsinki, Finland; Clinical and Chemical Pathology Department, National Cancer Institute, Cairo University, Cairo, Egypt; Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| | - Anu Loukola
- Helsinki Biobank, Helsinki University Hospital (HUS), Helsinki, Finland
| | - Yossra H S Zidi-Mouaffak
- Department of Pathology, University of Helsinki, Helsinki, Finland; Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Helsinki Biobank, Helsinki University Hospital (HUS), Helsinki, Finland
| | - Max Tamlander
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sanni Ruotsalainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Elina Kilpeläinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Nina Mars
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Clinicum, Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland; Broad Institute of MIT and Harvard, Cambridge, Massachusetts; Psychiatric and Neurodevelopmental Genetics Unit, Department of Psychiatry, Analytic and Translational Genetics Unit, Department of Medicine, and the Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts
| | - Kati Donner
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Olli Carpén
- Department of Pathology, University of Helsinki, Helsinki, Finland; Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Helsinki Biobank, Helsinki University Hospital (HUS), Helsinki, Finland
| |
Collapse
|
16
|
Casazza W, Inkster AM, Del Gobbo GF, Yuan V, Delahaye F, Marsit C, Park YP, Robinson WP, Mostafavi S, Dennis JK. Sex-dependent placental methylation quantitative trait loci provide insight into the prenatal origins of childhood onset traits and conditions. iScience 2024; 27:109047. [PMID: 38357671 PMCID: PMC10865402 DOI: 10.1016/j.isci.2024.109047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/19/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Molecular quantitative trait loci (QTLs) allow us to understand the biology captured in genome-wide association studies (GWASs). The placenta regulates fetal development and shows sex differences in DNA methylation. We therefore hypothesized that placental methylation QTL (mQTL) explain variation in genetic risk for childhood onset traits, and that effects differ by sex. We analyzed 411 term placentas from two studies and found 49,252 methylation (CpG) sites with mQTL and 2,489 CpG sites with sex-dependent mQTL. All mQTL were enriched in regions that typically affect gene expression in prenatal tissues. All mQTL were also enriched in GWAS results for growth- and immune-related traits, but male- and female-specific mQTL were more enriched than cross-sex mQTL. mQTL colocalized with trait loci at 777 CpG sites, with 216 (28%) specific to males or females. Overall, mQTL specific to male and female placenta capture otherwise overlooked variation in childhood traits.
Collapse
Affiliation(s)
- William Casazza
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital, Vancouver, BC, Canada
- Bioinformatics Graduate Program, University of British Columbia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
| | - Amy M. Inkster
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Giulia F. Del Gobbo
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Children’s Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Victor Yuan
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | | | - Carmen Marsit
- Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Yongjin P. Park
- Department of Statistics, University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wendy P. Robinson
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Sara Mostafavi
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital, Vancouver, BC, Canada
- Paul Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA
| | - Jessica K. Dennis
- Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital, Vancouver, BC, Canada
- Bioinformatics Graduate Program, University of British Columbia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
17
|
Minnai F, Biscarini F, Esposito M, Dragani TA, Bujanda L, Rahmouni S, Alarcón-Riquelme ME, Bernardo D, Carnero-Montoro E, Buti M, Zeberg H, Asselta R, Romero-Gómez M, Fernandez-Cadenas I, Fallerini C, Zguro K, Croci S, Baldassarri M, Bruttini M, Furini S, Renieri A, Colombo F. A genome-wide association study for survival from a multi-centre European study identified variants associated with COVID-19 risk of death. Sci Rep 2024; 14:3000. [PMID: 38321133 PMCID: PMC10847137 DOI: 10.1038/s41598-024-53310-x] [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: 11/20/2023] [Accepted: 01/30/2024] [Indexed: 02/08/2024] Open
Abstract
The clinical manifestations of SARS-CoV-2 infection vary widely among patients, from asymptomatic to life-threatening. Host genetics is one of the factors that contributes to this variability as previously reported by the COVID-19 Host Genetics Initiative (HGI), which identified sixteen loci associated with COVID-19 severity. Herein, we investigated the genetic determinants of COVID-19 mortality, by performing a case-only genome-wide survival analysis, 60 days after infection, of 3904 COVID-19 patients from the GEN-COVID and other European series (EGAS00001005304 study of the COVID-19 HGI). Using imputed genotype data, we carried out a survival analysis using the Cox model adjusted for age, age2, sex, series, time of infection, and the first ten principal components. We observed a genome-wide significant (P-value < 5.0 × 10-8) association of the rs117011822 variant, on chromosome 11, of rs7208524 on chromosome 17, approaching the genome-wide threshold (P-value = 5.19 × 10-8). A total of 113 variants were associated with survival at P-value < 1.0 × 10-5 and most of them regulated the expression of genes involved in immune response (e.g., CD300 and KLR genes), or in lung repair and function (e.g., FGF19 and CDH13). Overall, our results suggest that germline variants may modulate COVID-19 risk of death, possibly through the regulation of gene expression in immune response and lung function pathways.
Collapse
Affiliation(s)
- Francesca Minnai
- Institute of Biomedical Technologies, National Research Council, Via F.lli Cervi, 93, 20054, Segrate, MI, Italy
- Department of Medical Biotechnology and Translational Medicine (BioMeTra), Università degli Studi di Milano, Milan, Italy
| | - Filippo Biscarini
- Institute of Agricultural Biology and Biotechnology, National Research Council, Milan, Italy
| | - Martina Esposito
- Institute of Biomedical Technologies, National Research Council, Via F.lli Cervi, 93, 20054, Segrate, MI, Italy
| | | | - Luis Bujanda
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Biodonostia Health Research Institute, Universidad del País Vasco (UPV/EHU), San Sebastián, Spain
| | | | - Marta E Alarcón-Riquelme
- GENYO, University of Granada, Andalusian Regional Government, Granada, Spain
- Institute for Environmental Medicine, Karolinska Institute, Solna, Sweden
| | - David Bernardo
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Mucosal Immunology Lab, Unit of Excellence, Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid-CSIC, Valladolid, Spain
| | - Elena Carnero-Montoro
- GENYO, University of Granada, Andalusian Regional Government, Granada, Spain
- University of Granada, Granada, Spain
| | - Maria Buti
- Vall D'Hebron Institut de Recerca, Barcelona, Spain
| | - Hugo Zeberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Rosanna Asselta
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, MI, Italy
- IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Manuel Romero-Gómez
- Digestive Diseases Unit and CiberehdVirgen del Rocío University HospitalInstitute of Biomedicine of Seville (HUVR/CSIC/US), University of Seville, Seville, Spain
| | - Israel Fernandez-Cadenas
- Stroke Pharmacogenomics and Genetics Group, Sant Pau Hospital Research Institute, Barcelona, Spain
| | - Chiara Fallerini
- Medical Genetics, University of Siena, 53100, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, 53100, Siena, Italy
| | - Kristina Zguro
- Medical Genetics, University of Siena, 53100, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, 53100, Siena, Italy
| | - Susanna Croci
- Medical Genetics, University of Siena, 53100, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, 53100, Siena, Italy
| | - Margherita Baldassarri
- Medical Genetics, University of Siena, 53100, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, 53100, Siena, Italy
| | - Mirella Bruttini
- Medical Genetics, University of Siena, 53100, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, 53100, Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100, Siena, Italy
| | - Simone Furini
- Dipartimento di Ingegneria dell'Energia Elettrica e dell'Informazione "Guglielmo Marconi", Alma Mater Studiorum - Università di Bologna, Bologna, Italy
| | - Alessandra Renieri
- Medical Genetics, University of Siena, 53100, Siena, Italy
- Department of Medical Biotechnologies, Med Biotech Hub and Competence Center, University of Siena, 53100, Siena, Italy
- Genetica Medica, Azienda Ospedaliero-Universitaria Senese, 53100, Siena, Italy
| | - Francesca Colombo
- Institute of Biomedical Technologies, National Research Council, Via F.lli Cervi, 93, 20054, Segrate, MI, Italy.
| |
Collapse
|
18
|
Weckman MJ, Karikoski NP, Raekallio MR, Box JR, Kvist L. Genome-wide association study suggests genetic candidate loci of insulin dysregulation in Finnhorses. Vet J 2024; 303:106063. [PMID: 38232813 DOI: 10.1016/j.tvjl.2024.106063] [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: 09/06/2022] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 01/19/2024]
Abstract
Equine metabolic syndrome (EMS) is a common welfare problem in horses worldwide. It is characterized by insulin dysregulation (ID), predisposition to laminitis and often obesity. EMS is multifactorial by nature, with both the environment and genetics contributing to the phenotype. Environmental factors, such as feeding and exercise, can be controlled, thus forming the basis for treatment and prevention. Genetic factors, by contrast, are less well-known and not easily controllable. The aim of this study was to identify potential genetic loci influencing ID/EMS in Finnhorses. A single-breed (Finnhorse) case-control genome-wide association study (GWAS) of ID was conducted with controls that included age-appropriate non-ID horses. ID status was determined with an oral sugar test (OST) for fasted horses. Seventy-one Finnhorses participated (n = 34 ID, n = 37 control). DNA samples (hair roots) were genotyped for 65 157 single-nucleotide polymorphisms (SNPs) with the Illumina Equine SNP70 BeadChip, and these data were analysed for association and FST outliers with genomic tools. P-values that exceeded the suggestive threshold (P = 1.00 ×10-5) were found in SNP BIEC2_383954 (P = 3.45 ×10-6) in chromosome 17 and SNP BIEC2_312374 (P = 1.89 ×10-5) in chromosome 15. Hierarchical and Bayesian FST outlier tests also detected these SNPs. Potential candidate genes associated with the ID close to SNP BIEC2_383954, with functions in carbohydrate metabolism, were Arginine and Glutamate Rich 1 (ARGLU1) and Ephrin-B2 (EFNB2).
Collapse
Affiliation(s)
- M J Weckman
- Department of Equine and Small Animal Sciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland.
| | - N P Karikoski
- Department of Equine and Small Animal Sciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - M R Raekallio
- Department of Equine and Small Animal Sciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - J R Box
- Department of Equine and Small Animal Sciences, Faculty of Veterinary Medicine, University of Helsinki, P.O. Box 57, FI-00014 Helsinki, Finland
| | - L Kvist
- Ecology and Genetics Research Unit, University of Oulu, P.O. Box 8000, FI-3000 Oulu, Finland
| |
Collapse
|
19
|
Halvorsen M, de Schipper E, Boberg J, Strom N, Hagen K, Lindblad-Toh K, Karlsson E, Pedersen N, Bulik C, Fundín B, Landén M, Kvale G, Hansen B, Haavik J, Mattheisen M, Rück C, Mataix-Cols D, Crowley J. A Burden of Rare Copy Number Variants in Obsessive-Compulsive Disorder. RESEARCH SQUARE 2024:rs.3.rs-3749504. [PMID: 38260575 PMCID: PMC10802697 DOI: 10.21203/rs.3.rs-3749504/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Current genetic research on obsessive-compulsive disorder (OCD) supports contributions to risk specifically from common single nucleotide variants (SNVs), along with rare coding SNVs and small insertion-deletions (indels). The contribution to OCD risk from large, rare copy number variants (CNVs), however, has not been formally assessed at a similar scale. Here we describe an analysis of rare CNVs called from genotype array data in 2,248 deeply phenotyped OCD cases and 3,608 unaffected controls from Sweden and Norway. We found that in general cases carry an elevated burden of large (>30kb, at least 15 probes) CNVs (OR=1.12, P=1.77×10-3). The excess rate of these CNVs in cases versus controls was around 0.07 (95% CI 0.02-0.11, P=2.58×10-3). This signal was largely driven by CNVs overlapping protein-coding regions (OR=1.19, P=3.08×10-4), particularly deletions impacting loss-of-function intolerant genes (pLI>0.995, OR=4.12, P=2.54×10-5). We did not identify any specific locus where CNV burden was associated with OCD case status at genome-wide significance, but we noted non-random recurrence of CNV deletions in cases (permutation P = 2.60×10-3). In cases where sufficient clinical data were available (n=1612) we found that carriers of neurodevelopmental duplications were more likely to have comorbid autism (P<0.001), and that carriers of deletions overlapping neurodevelopmental genes had lower treatment response (P=0.02). The results demonstrate a contribution of large, rare CNVs to OCD risk, and suggest that studies of rare coding variation in OCD would have increased power to identify risk genes if this class of variation were incorporated into formal tests.
Collapse
|
20
|
Latham KE. Preimplantation genetic testing: A remarkable history of pioneering, technical challenges, innovations, and ethical considerations. Mol Reprod Dev 2024; 91:e23727. [PMID: 38282313 DOI: 10.1002/mrd.23727] [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: 10/11/2023] [Accepted: 12/15/2023] [Indexed: 01/30/2024]
Abstract
Preimplantation genetic testing (PGT) has emerged as a powerful companion to assisted reproduction technologies. The origins and history of PGT are reviewed here, along with descriptions of advances in molecular assays and sampling methods, their capabilities, and their applications in preventing genetic diseases and enhancing pregnancy outcomes. Additionally, the potential for increasing accuracy and genome coverage is considered, as well as some of the emerging ethical and legislative considerations related to the expanding capabilities of PGT.
Collapse
Affiliation(s)
- Keith E Latham
- Department of Animal Science, Michigan State University, East Lansing, Michigan, USA
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, Michigan, USA
- Reproductive and Developmental Sciences Program, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
21
|
Nanjala R, Mbiyavanga M, Hashim S, de Villiers S, Mulder N. Assessing HLA imputation accuracy in a West African population. PLoS One 2023; 18:e0291437. [PMID: 37768905 PMCID: PMC10538777 DOI: 10.1371/journal.pone.0291437] [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: 05/12/2023] [Accepted: 08/28/2023] [Indexed: 09/30/2023] Open
Abstract
The Human Leukocyte Antigen (HLA) region plays an important role in autoimmune and infectious diseases. HLA is a highly polymorphic region and thus difficult to impute. We, therefore, sought to evaluate HLA imputation accuracy, specifically in a West African population, since they are understudied and are known to harbor high genetic diversity. The study sets were selected from 315 Gambian individuals within the Gambian Genome Variation Project (GGVP) Whole Genome Sequence datasets. Two different arrays, Illumina Omni 2.5 and Human Hereditary and Health in Africa (H3Africa), were assessed for the appropriateness of their markers, and these were used to test several imputation panels and tools. The reference panels were chosen from the 1000 Genomes (1kg-All), 1000 Genomes African (1kg-Afr), 1000 Genomes Gambian (1kg-Gwd), H3Africa, and the HLA Multi-ethnic datasets. HLA-A, HLA-B, and HLA-C alleles were imputed using HIBAG, SNP2HLA, CookHLA, and Minimac4, and concordance rate was used as an assessment metric. The best performing tool was found to be HIBAG, with a concordance rate of 0.84, while the best performing reference panel was the H3Africa panel, with a concordance rate of 0.62. Minimac4 (0.75) was shown to increase HLA-B allele imputation accuracy compared to HIBAG (0.71), SNP2HLA (0.51) and CookHLA (0.17). The H3Africa and Illumina Omni 2.5 array performances were comparable, showing that genotyping arrays have less influence on HLA imputation in West African populations. The findings show that using a larger population-specific reference panel and the HIBAG tool improves the accuracy of HLA imputation in a West African population.
Collapse
Affiliation(s)
- Ruth Nanjala
- Department of Biochemistry and Biotechnology, Pwani University, Kilifi, Kenya
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, University of Cape Town, Cape Town, South Africa
| | - Mamana Mbiyavanga
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, University of Cape Town, Cape Town, South Africa
| | - Suhaila Hashim
- Department of Biochemistry and Biotechnology, Pwani University, Kilifi, Kenya
- Pwani University Biosciences Research Centre, Pwani University, Kilifi, Kenya
| | - Santie de Villiers
- Department of Biochemistry and Biotechnology, Pwani University, Kilifi, Kenya
- Pwani University Biosciences Research Centre, Pwani University, Kilifi, Kenya
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
22
|
Sakaue S, Gurajala S, Curtis M, Luo Y, Choi W, Ishigaki K, Kang JB, Rumker L, Deutsch AJ, Schönherr S, Forer L, LeFaive J, Fuchsberger C, Han B, Lenz TL, de Bakker PIW, Okada Y, Smith AV, Raychaudhuri S. Tutorial: a statistical genetics guide to identifying HLA alleles driving complex disease. Nat Protoc 2023; 18:2625-2641. [PMID: 37495751 PMCID: PMC10786448 DOI: 10.1038/s41596-023-00853-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 04/27/2023] [Indexed: 07/28/2023]
Abstract
The human leukocyte antigen (HLA) locus is associated with more complex diseases than any other locus in the human genome. In many diseases, HLA explains more heritability than all other known loci combined. In silico HLA imputation methods enable rapid and accurate estimation of HLA alleles in the millions of individuals that are already genotyped on microarrays. HLA imputation has been used to define causal variation in autoimmune diseases, such as type I diabetes, and in human immunodeficiency virus infection control. However, there are few guidelines on performing HLA imputation, association testing, and fine mapping. Here, we present a comprehensive tutorial to impute HLA alleles from genotype data. We provide detailed guidance on performing standard quality control measures for input genotyping data and describe options to impute HLA alleles and amino acids either locally or using the web-based Michigan Imputation Server, which hosts a multi-ancestry HLA imputation reference panel. We also offer best practice recommendations to conduct association tests to define the alleles, amino acids, and haplotypes that affect human traits. Along with the pipeline, we provide a step-by-step online guide with scripts and available software ( https://github.com/immunogenomics/HLA_analyses_tutorial ). This tutorial will be broadly applicable to large-scale genotyping data and will contribute to defining the role of HLA in human diseases across global populations.
Collapse
Affiliation(s)
- Saori Sakaue
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Saisriram Gurajala
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michelle Curtis
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yang Luo
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Wanson Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Kazuyoshi Ishigaki
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Joyce B Kang
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Laurie Rumker
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Aaron J Deutsch
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Program in Metabolism, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas Forer
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Jonathon LeFaive
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Christian Fuchsberger
- Institute of Genetic Epidemiology, Department of Genetics, Medical University of Innsbruck, Innsbruck, Austria
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Institute for Biomedicine, Eurac Research, Bolzano, Italy
| | - Buhm Han
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, South Korea
| | - Tobias L Lenz
- Research Unit for Evolutionary Immunogenomics, Department of Biology, University of Hamburg, Hamburg, Germany
| | - Paul I W de Bakker
- Data and Computational Sciences, Vertex Pharmaceuticals, Boston, MA, USA
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Center for Infectious Disease Education and Research (CiDER), Osaka University, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Albert V Smith
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Divisions of Genetics and Rheumatology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Centre for Genetics and Genomics Versus Arthritis, University of Manchester, Manchester, UK.
| |
Collapse
|
23
|
Yuan S, Li X, Liu Q, Wang Z, Jiang X, Burgess S, Larsson SC. Physical Activity, Sedentary Behavior, and Type 2 Diabetes: Mendelian Randomization Analysis. J Endocr Soc 2023; 7:bvad090. [PMID: 37415875 PMCID: PMC10321115 DOI: 10.1210/jendso/bvad090] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Indexed: 07/08/2023] Open
Abstract
Context The causality and pathways of the associations between physical activity and inactivity and the risk of type 2 diabetes remain inconclusive. Objective We conducted an updated mendelian randomization (MR) study to explore the associations of moderate-to-vigorous physical activity (MVPA) and leisure screen time (LST) with type 2 diabetes mellitus (T2DM). Methods Genetic variants strongly associated with MVPA or LST with low linkage disequilibrium were selected as instrumental variables from a genome-wide meta-analysis including more than 600 000 individuals. Summary-level data on T2DM were obtained from the DIAbetes Genetics Replication And Meta-analysis consortium including 898 130 individuals. Data on possible intermediates (adiposity indicators, lean mass, glycemic traits, and inflammatory biomarkers) were extracted from large-scale genome-wide association studies (n = 21 758-681 275). Univariable and multivariable MR analyses were performed to estimate the total and direct effects of MVPA and LST on T2DM. Methylation MR analysis was performed for MVPA in relation to diabetes. Results The odds ratio of T2DM was 0.70 (95% CI, 0.55-0.88; P = .002) per unit increase in the log-odds ratio of having MVPA and 1.45 (95% CI, 1.30-1.62; P = 7.62 × 10-11) per SD increase in genetically predicted LST. These associations attenuated in multivariable MR analyses adjusted for genetically predicted waist-to-hip ratio, body mass index, lean mass, and circulating C-reactive protein. The association between genetically predicted MVPA and T2DM attenuated after adjusting for genetically predicted fasting insulin levels. Two physical activity-related methylation biomarkers (cg17332422 in ADAMTS2 and cg09531019) were associated with the risk of T2DM (P < .05). Conclusion The study suggests causal associations of MVPA and LST with T2DM that appear to be mediated by obesity, lean mass, and chronic low-grade inflammation.
Collapse
Affiliation(s)
- Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Xue Li
- School of Public Health and the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, China
| | - Qianwen Liu
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Zhe Wang
- Icahn School of Medicine at Mount Sinai, The Charles Bronfman Institute for Personalized Medicine, New York, NY 10029, USA
| | - Xia Jiang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 1TN, UK
- Department of Public Health and Primary Care, University of Cambridge, Cambridge CB2 1TN, UK
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, 17165, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, 75185, Sweden
| |
Collapse
|
24
|
Berghout BP, Bos D, Koudstaal PJ, Ikram MA, Ikram MK. Risk of recurrent stroke in Rotterdam between 1990 and 2020: a population-based cohort study. THE LANCET REGIONAL HEALTH. EUROPE 2023; 30:100651. [PMID: 37228392 PMCID: PMC10205482 DOI: 10.1016/j.lanepe.2023.100651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/27/2023]
Abstract
Background After an initial stroke, current clinical practice is aimed at preventing recurrent stroke. Thus far, population-based estimates on the risk of recurrent stroke remain scarce. Here we describe the risk of recurrent stroke in a population-based cohort study. Methods We included Rotterdam Study participants who developed a first-ever incident stroke during follow-up between 1990 until 2020. During further follow-up, these participants were monitored for the occurrence of a recurrent stroke. We determined stroke subtypes based on clinical and imaging information. We calculated ten-year overall and sex-specific cumulative incidences of first recurrent stroke. To reflect changing secondary preventive strategies employed in recent decades, we then calculated the risk of recurrent stroke within ten-year epochs based on first-ever stroke date (1990-2000, 2000-2010 and 2010-2020). Findings In total, 1701 participants (mean age 80.3 years, 59.8% women) from 14,163 community-living individuals suffered a first stroke between 1990 and 2020. Of these strokes, 1111 (65.3%) were ischaemic, 141 (8.3%) haemorrhagic, and 449 (26.4%) unspecified. During 6585.3 person-years of follow-up, 331 (19.5%) suffered a recurrent stroke, of which 178 (53.8%) were ischaemic, 34 (10.3%) haemorrhagic and 119 (36.0%) unspecified. Median time between first and recurrent stroke was 1.8 (interquartile range 0.5-4.6) years. Overall ten-year recurrence risk following first-ever stroke was 18.0% (95% CI 16.2%-19.8%), 19.3% (16.3%-22.3%) in men and 17.1% (14.8%-19.4%) in women. Recurrent stroke risk declined over time, with a ten-year risk of 21.4% (17.9%-24.9%) between 1990 and 2000 and 11.0% (8.3%-13.8%) between 2010 and 2020. Interpretation In this population-based study, almost one in five people with first-ever stroke suffered a recurrence within ten years of the initial stroke. Furthermore, recurrence risk declined between 2010 and 2020. Funding Netherlands Organization for Health Research and Development, EU's Horizon 2020 research programme and the Erasmus Medical Centre MRACE grant.
Collapse
Affiliation(s)
- Bernhard P. Berghout
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, Netherlands
- Department of Neurology, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - Peter J. Koudstaal
- Department of Neurology, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, Netherlands
| | - M. Kamran Ikram
- Department of Epidemiology, Erasmus University Medical Centre, Rotterdam, Netherlands
- Department of Neurology, Erasmus University Medical Centre, Rotterdam, Netherlands
| |
Collapse
|
25
|
Singh P, Crossman DK, Zhou L, Wang X, Sharafeldin N, Hageman L, Blanco JG, Burridge PW, Armenian SH, Balis FM, Hawkins DS, Keller FG, Hudson MM, Neglia JP, Ritchey AK, Ginsberg JP, Landier W, Bhatia S. Haptoglobin Gene Expression and Anthracycline-Related Cardiomyopathy in Childhood Cancer Survivors: A COG-ALTE03N1 Report. JACC CardioOncol 2023; 5:392-401. [PMID: 37397079 PMCID: PMC10308004 DOI: 10.1016/j.jaccao.2022.09.009] [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: 06/13/2022] [Revised: 09/27/2022] [Accepted: 09/30/2022] [Indexed: 02/10/2023] Open
Abstract
Background Anthracycline-related cardiomyopathy is a leading cause of premature death in childhood cancer survivors. The high interindividual variability in risk suggests the need to understand the underlying pathogenesis. Objectives The authors interrogated differentially expressed genes (DEGs) to identify genetic variants serving regulatory functions or genetic variants not easily identified when using genomewide array platforms. Using leads from DEGs, candidate copy number variants (CNVs) and single-nucleotide variants (SNVs) were genotyped. Methods Messenger RNA sequencing was performed on total RNA from peripheral blood of 40 survivors with cardiomyopathy (cases) and 64 matched survivors without cardiomyopathy (control subjects). Conditional logistic regression analysis adjusting for sex, age at cancer diagnosis, anthracycline dose, and chest radiation was used to assess the associations between gene expression and cardiomyopathy and between CNVs and SNVs and cardiomyopathy. Results Haptoglobin (HP) was identified as the top DEG. Participants with higher HP gene expression had 6-fold greater odds of developing cardiomyopathy (OR: 6.4; 95% CI: 1.4-28.6). The HP2-specific allele among the HP genotypes (HP1-1, HP1-2, and HP2-2) had higher transcript levels, as did the G allele among SNVs previously reported to be associated with HP gene expression (rs35283911 and rs2000999). The HP1-2 and HP2-2 genotypes combined with the G/G genotype for rs35283911 and/or rs2000999 placed the survivors at 4-fold greater risk (OR: 3.9; 95% CI: 1.0-14.5) for developing cardiomyopathy. Conclusions These findings provide evidence of a novel association between HP2 allele and cardiomyopathy. HP binds to free hemoglobin to form an HP-hemoglobin complex, thereby preventing oxidative damage from free heme iron, thus providing biological plausibility to the mechanistic basis of the present observation.
Collapse
Affiliation(s)
- Purnima Singh
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - David K. Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Liting Zhou
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xuexia Wang
- Department of Mathematics, University of North Texas, Denton, Texas, USA
| | - Noha Sharafeldin
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lindsey Hageman
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Javier G. Blanco
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Paul W. Burridge
- Department of Pharmacology, Northwestern University, Chicago, Illinois, USA
| | - Saro H. Armenian
- Department of Population Sciences, City of Hope, Duarte, California
| | - Frank M. Balis
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | - Frank G. Keller
- Children’s Healthcare of Atlanta, Emory University, Atlanta, Georgia, USA
| | | | | | - A. Kim Ritchey
- Children’s Hospital of Pittsburgh of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Jill P. Ginsberg
- Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
26
|
Kabbani D, Akika R, Wahid A, Daly AK, Cascorbi I, Zgheib NK. Pharmacogenomics in practice: a review and implementation guide. Front Pharmacol 2023; 14:1189976. [PMID: 37274118 PMCID: PMC10233068 DOI: 10.3389/fphar.2023.1189976] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Considerable efforts have been exerted to implement Pharmacogenomics (PGx), the study of interindividual variations in DNA sequence related to drug response, into routine clinical practice. In this article, we first briefly describe PGx and its role in improving treatment outcomes. We then propose an approach to initiate clinical PGx in the hospital setting. One should first evaluate the available PGx evidence, review the most relevant drugs, and narrow down to the most actionable drug-gene pairs and related variant alleles. This is done based on data curated and evaluated by experts such as the pharmacogenomics knowledge implementation (PharmGKB) and the Clinical Pharmacogenetics Implementation Consortium (CPIC), as well as drug regulatory authorities such as the US Food and Drug Administration (FDA) and European Medicinal Agency (EMA). The next step is to differentiate reactive point of care from preemptive testing and decide on the genotyping strategy being a candidate or panel testing, each of which has its pros and cons, then work out the best way to interpret and report PGx test results with the option of integration into electronic health records and clinical decision support systems. After test authorization or testing requirements by the government or drug regulators, putting the plan into action involves several stakeholders, with the hospital leadership supporting the process and communicating with payers, the pharmacy and therapeutics committee leading the process in collaboration with the hospital laboratory and information technology department, and healthcare providers (HCPs) ordering the test, understanding the results, making the appropriate therapeutic decisions, and explaining them to the patient. We conclude by recommending some strategies to further advance the implementation of PGx in practice, such as the need to educate HCPs and patients, and to push for more tests' reimbursement. We also guide the reader to available PGx resources and examples of PGx implementation programs and initiatives.
Collapse
Affiliation(s)
- Danya Kabbani
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Reem Akika
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ahmed Wahid
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Ann K. Daly
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ingolf Cascorbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nathalie Khoueiry Zgheib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
27
|
Reddi HV, Wand H, Funke B, Zimmermann MT, Lebo MS, Qian E, Shirts BH, Zou YS, Zhang BM, Rose NC, Abu-El-Haija A. Laboratory perspectives in the development of polygenic risk scores for disease: A points to consider statement of the American College of Medical Genetics and Genomics (ACMG). Genet Med 2023; 25:100804. [PMID: 36971772 DOI: 10.1016/j.gim.2023.100804] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 03/29/2023] Open
Affiliation(s)
- Honey V Reddi
- Department of Pathology & Laboratory Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Hannah Wand
- Division of Cardiovascular Medicine, Department of Medicine, Stanford Medicine, Stanford, CA
| | | | - Michael T Zimmermann
- Bioinformatics Research and Development Laboratory, Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Matthew S Lebo
- Laboratory for Molecular Medicine, Mass General Brigham, Cambridge, MA
| | - Emily Qian
- Department of Genetics, Yale University, New Haven, CT
| | - Brian H Shirts
- Department of Laboratory Medicine & Pathology, UW Medicine, University of Washington, Seattle, WA
| | - Ying S Zou
- Department of Genomic Medicine and Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Bing M Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA
| | - Nancy C Rose
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah Health, Salt Lake City, UT
| | - Aya Abu-El-Haija
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA; Harvard Medical School, Boston, MA
| |
Collapse
|
28
|
Why does the X chromosome lag behind autosomes in GWAS findings? PLoS Genet 2023; 19:e1010472. [PMID: 36848382 PMCID: PMC9997976 DOI: 10.1371/journal.pgen.1010472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/09/2023] [Accepted: 02/15/2023] [Indexed: 03/01/2023] Open
Abstract
The X-chromosome is among the largest human chromosomes. It differs from autosomes by a number of important features including hemizygosity in males, an almost complete inactivation of one copy in females, and unique patterns of recombination. We used data from the Catalog of Published Genome Wide Association Studies to compare densities of the GWAS-detected SNPs on the X-chromosome and autosomes. The density of GWAS-detected SNPs on the X-chromosome is 6-fold lower compared to the density of the GWAS-detected SNPs on autosomes. Differences between the X-chromosome and autosomes cannot be explained by differences in the overall SNP density, lower X-chromosome coverage by genotyping platforms or low call rate of X-chromosomal SNPs. Similar differences in the density of GWAS-detected SNPs were found in female-only GWASs (e.g. ovarian cancer GWASs). We hypothesized that the lower density of GWAS-detected SNPs on the X-chromosome compared to autosomes is not a result of a methodological bias, e.g. differences in coverage or call rates, but has a real underlying biological reason-a lower density of functional SNPs on the X-chromosome versus autosomes. This hypothesis is supported by the observation that (i) the overall SNP density of X-chromosome is lower compared to the SNP density on autosomes and that (ii) the density of genic SNPs on the X-chromosome is lower compared to autosomes while densities of intergenic SNPs are similar.
Collapse
|
29
|
Nanjala R, Mbiyavanga M, Hashim S, de Villiers S, Mulder N. Assessing HLA imputation accuracy in a West African population. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.23.525129. [PMID: 36747714 PMCID: PMC9900754 DOI: 10.1101/2023.01.23.525129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The Human Leukocyte Antigen (HLA) region plays an important role in autoimmune and infectious diseases. HLA is a highly polymorphic region and thus difficult to impute. We therefore sought to evaluate HLA imputation accuracy, specifically in a West African population, since they are understudied and are known to harbor high genetic diversity. The study sets were selected from Gambian individuals within the Gambian Genome Variation Project (GGVP) Whole Genome Sequence datasets. Two different arrays, Illumina Omni 2.5 and Human Hereditary and Health in Africa (H3Africa), were assessed for the appropriateness of their markers, and these were used to test several imputation panels and tools. The reference panels were chosen from the 1000 Genomes dataset (1kg-All), 1000 Genomes African dataset (1kg-Afr), 1000 Genomes Gambian dataset (1kg-Gwd), H3Africa dataset and the HLA Multi-ethnic dataset. HLA-A, HLA-B and HLA-C alleles were imputed using HIBAG, SNP2HLA, CookHLA and Minimac4, and concordance rate was used as an assessment metric. Overall, the best performing tool was found to be HIBAG, with a concordance rate of 0.84, while the best performing reference panel was the H3Africa panel with a concordance rate of 0.62. Minimac4 (0.75) was shown to increase HLA-B allele imputation accuracy compared to HIBAG (0.71), SNP2HLA (0.51) and CookHLA (0.17). The H3Africa and Illumina Omni 2.5 array performances were comparable, showing that genotyping arrays have less influence on HLA imputation in West African populations. The findings show that using a larger population-specific reference panel and the HIBAG tool improves the accuracy of HLA imputation in West African populations. Author Summary For studies that associate a particular HLA type to a phenotypic trait for instance HIV susceptibility or control, genotype imputation remains the main method for acquiring a larger sample size. Genotype imputation, process of inferring unobserved genotypes, is a statistical technique and thus deals with probabilities. Also, the HLA region is highly variable and therefore difficult to impute. In view of this, it is important to assess HLA imputation accuracy especially in African populations. This is because the African genome has high diversity, and such studies have hardly been conducted in African populations. This work highlights that using HIBAG imputation tool and a larger population-specific reference panel increases HLA imputation accuracy in an African population.
Collapse
Affiliation(s)
- Ruth Nanjala
- Department of Biochemistry and Biotechnology, Pwani University, Kenya
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, University of Cape Town, South Africa
| | - Mamana Mbiyavanga
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, University of Cape Town, South Africa
| | - Suhaila Hashim
- Department of Biochemistry and Biotechnology, Pwani University, Kenya
- Pwani University Biosciences Research Centre, Pwani University, Kenya
| | - Santie de Villiers
- Department of Biochemistry and Biotechnology, Pwani University, Kenya
- Pwani University Biosciences Research Centre, Pwani University, Kenya
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, IDM, University of Cape Town, South Africa
| |
Collapse
|
30
|
Next generation sequencing technologies to explore the diversity of germplasm resources: achievements and trends in tomato. Comput Struct Biotechnol J 2022; 20:6250-6258. [PMID: 36420160 PMCID: PMC9676195 DOI: 10.1016/j.csbj.2022.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/10/2022] [Accepted: 11/10/2022] [Indexed: 11/14/2022] Open
Abstract
Tomato is one of the major vegetable crops grown worldwide and a model species for genetic and biological research. Progress in genomic technologies made possible the development of forefront methods for high-scale sequencing, providing comprehensive insight into the genetic architecture of germplasm resources. This review revisits next-generation sequencing strategies and applications to investigate the diversity of tomato, describing the common platforms used for SNP genotyping of large collections, de novo sequencing, and whole genome resequencing. Significant findings in evolutionary history are outlined, thus discussing how genomics has provided new hints about the processes behind domestication. Finally, achievement and perspectives on pan-genome construction and graphical pan-genome development toward precise mining of the natural variation to be exploited for breeding purposes are presented.
Collapse
|
31
|
Kim KW, Nawade B, Nam J, Chu SH, Ha J, Park YJ. Development of an inclusive 580K SNP array and its application for genomic selection and genome-wide association studies in rice. FRONTIERS IN PLANT SCIENCE 2022; 13:1036177. [PMID: 36352876 PMCID: PMC9637963 DOI: 10.3389/fpls.2022.1036177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 09/30/2022] [Indexed: 06/16/2023]
Abstract
Rice is a globally cultivated crop and is primarily a staple food source for more than half of the world's population. Various single-nucleotide polymorphism (SNP) arrays have been developed and utilized as standard genotyping methods for rice breeding research. Considering the importance of SNP arrays with more inclusive genetic information for GWAS and genomic selection, we integrated SNPs from eight different data resources: resequencing data from the Korean World Rice Collection (KRICE) of 475 accessions, 3,000 rice genome project (3 K-RGP) data, 700 K high-density rice array, Affymetrix 44 K SNP array, QTARO, Reactome, and plastid and GMO information. The collected SNPs were filtered and selected based on the breeder's interest, covering all key traits or research areas to develop an integrated array system representing inclusive genomic polymorphisms. A total of 581,006 high-quality SNPs were synthesized with an average distance of 200 bp between adjacent SNPs, generating a 580 K Axiom Rice Genotyping Chip (580 K _ KNU chip). Further validation of this array on 4,720 genotypes revealed robust and highly efficient genotyping. This has also been demonstrated in genome-wide association studies (GWAS) and genomic selection (GS) of three traits: clum length, heading date, and panicle length. Several SNPs significantly associated with cut-off, -log10 p-value >7.0, were detected in GWAS, and the GS predictabilities for the three traits were more than 0.5, in both rrBLUP and convolutional neural network (CNN) models. The Axiom 580 K Genotyping array will provide a cost-effective genotyping platform and accelerate rice GWAS and GS studies.
Collapse
Affiliation(s)
- Kyu-Won Kim
- Center for Crop Breeding on Omics and Artificial Intelligence, Kongju National University, Yesan, South Korea
| | - Bhagwat Nawade
- Center for Crop Breeding on Omics and Artificial Intelligence, Kongju National University, Yesan, South Korea
| | - Jungrye Nam
- Center for Crop Breeding on Omics and Artificial Intelligence, Kongju National University, Yesan, South Korea
| | - Sang-Ho Chu
- Center for Crop Breeding on Omics and Artificial Intelligence, Kongju National University, Yesan, South Korea
| | - Jungmin Ha
- Department of Plant Science, Gangneung-Wonju National University, Gangneung, South Korea
| | - Yong-Jin Park
- Center for Crop Breeding on Omics and Artificial Intelligence, Kongju National University, Yesan, South Korea
- Department of Plant Resources, College of Industrial Sciences, Kongju National University, Yesan, South Korea
| |
Collapse
|
32
|
Nguyen DT, Tran TTH, Tran MH, Tran K, Pham D, Duong NT, Nguyen Q, Vo NS. A comprehensive evaluation of polygenic score and genotype imputation performances of human SNP arrays in diverse populations. Sci Rep 2022; 12:17556. [PMID: 36266455 PMCID: PMC9585077 DOI: 10.1038/s41598-022-22215-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/11/2022] [Indexed: 01/13/2023] Open
Abstract
Regardless of the overwhelming use of next-generation sequencing technologies, microarray-based genotyping combined with the imputation of untyped variants remains a cost-effective means to interrogate genetic variations across the human genome. This technology is widely used in genome-wide association studies (GWAS) at bio-bank scales, and more recently, in polygenic score (PGS) analysis to predict and stratify disease risk. Over the last decade, human genotyping arrays have undergone a tremendous growth in both number and content making a comprehensive evaluation of their performances became more important. Here, we performed a comprehensive performance assessment for 23 available human genotyping arrays in 6 ancestry groups using diverse public and in-house datasets. The analyses focus on performance estimation of derived imputation (in terms of accuracy and coverage) and PGS (in terms of concordance to PGS estimated from whole-genome sequencing data) in three different traits and diseases. We found that the arrays with a higher number of SNPs are not necessarily the ones with higher imputation performance, but the arrays that are well-optimized for the targeted population could provide very good imputation performance. In addition, PGS estimated by imputed SNP array data is highly correlated to PGS estimated by whole-genome sequencing data in most cases. When optimal arrays are used, the correlations of PGS between two types of data are higher than 0.97, but interestingly, arrays with high density can result in lower PGS performance. Our results suggest the importance of properly selecting a suitable genotyping array for PGS applications. Finally, we developed a web tool that provides interactive analyses of tag SNP contents and imputation performance based on population and genomic regions of interest. This study would act as a practical guide for researchers to design their genotyping arrays-based studies. The tool is available at: https://genome.vinbigdata.org/tools/saa/ .
Collapse
Affiliation(s)
- Dat Thanh Nguyen
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Vietnam.
- Faculty of Biosciences, Norwegian University of Life Sciences, Ås, Norway.
| | - Trang T H Tran
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Vietnam
- GeneStory JSC, Hanoi, Vietnam
| | - Mai Hoang Tran
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Vietnam
- GeneStory JSC, Hanoi, Vietnam
| | - Khai Tran
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Vietnam
| | - Duy Pham
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Nguyen Thuy Duong
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Vietnam
- GeneStory JSC, Hanoi, Vietnam
- Institute of Genome Research, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Quan Nguyen
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia.
| | - Nam S Vo
- Center for Biomedical Informatics, Vingroup Big Data Institute, Hanoi, Vietnam.
- GeneStory JSC, Hanoi, Vietnam.
| |
Collapse
|
33
|
Sellers R, Riglin L, Harold GT, Thapar A. Using genetic designs to identify likely causal environmental contributions to psychopathology. Dev Psychopathol 2022; 34:1-13. [PMID: 36200346 DOI: 10.1017/s0954579422000906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The multifactorial nature of psychopathology, whereby both genetic and environmental factors contribute risk, has long been established. In this paper, we provide an update on genetically informative designs that are utilized to disentangle genetic and environmental contributions to psychopathology. We provide a brief reminder of quantitative behavioral genetic research designs that have been used to identify potentially causal environmental processes, accounting for genetic contributions. We also provide an overview of recent molecular genetic approaches that utilize genome-wide association study data which are increasingly being applied to questions relevant to psychopathology research. While genetically informative designs typically have been applied to investigate the origins of psychopathology, we highlight how these approaches can also be used to elucidate potential causal environmental processes that contribute to developmental course and outcomes. We highlight the need to use genetically sensitive designs that align with intervention and prevention science efforts, by considering strengths-based environments to investigate how positive environments can mitigate risk and promote children's strengths.
Collapse
Affiliation(s)
- Ruth Sellers
- Brighton & Sussex Medical School, University of Sussex, Brighton, UK
| | - Lucy Riglin
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Wolfson Centre for Young People's Mental Health, Cardiff University, Cardiff, UK
| | - Gordon T Harold
- Faculty of Education, University of Cambridge, Cambridge, UK
- School of Medicine, Child and Adolescent Psychiatry Unit, University College Dublin, Dublin, Ireland
| | - Anita Thapar
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, UK
- Wolfson Centre for Young People's Mental Health, Cardiff University, Cardiff, UK
| |
Collapse
|
34
|
Hanks SC, Forer L, Schönherr S, LeFaive J, Martins T, Welch R, Gagliano Taliun SA, Braff D, Johnsen JM, Kenny EE, Konkle BA, Laakso M, Loos RF, McCarroll S, Pato C, Pato MT, Smith AV, Boehnke M, Scott LJ, Fuchsberger C. Extent to which array genotyping and imputation with large reference panels approximate deep whole-genome sequencing. Am J Hum Genet 2022; 109:1653-1666. [PMID: 35981533 PMCID: PMC9502057 DOI: 10.1016/j.ajhg.2022.07.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 07/20/2022] [Indexed: 01/02/2023] Open
Abstract
Understanding the genetic basis of human diseases and traits is dependent on the identification and accurate genotyping of genetic variants. Deep whole-genome sequencing (WGS), the gold standard technology for SNP and indel identification and genotyping, remains very expensive for most large studies. Here, we quantify the extent to which array genotyping followed by genotype imputation can approximate WGS in studies of individuals of African, Hispanic/Latino, and European ancestry in the US and of Finnish ancestry in Finland (a population isolate). For each study, we performed genotype imputation by using the genetic variants present on the Illumina Core, OmniExpress, MEGA, and Omni 2.5M arrays with the 1000G, HRC, and TOPMed imputation reference panels. Using the Omni 2.5M array and the TOPMed panel, ≥90% of bi-allelic single-nucleotide variants (SNVs) are well imputed (r2 > 0.8) down to minor-allele frequencies (MAFs) of 0.14% in African, 0.11% in Hispanic/Latino, 0.35% in European, and 0.85% in Finnish ancestries. There was little difference in TOPMed-based imputation quality among the arrays with >700k variants. Individual-level imputation quality varied widely between and within the three US studies. Imputation quality also varied across genomic regions, producing regions where even common (MAF > 5%) variants were consistently not well imputed across ancestries. The extent to which array genotyping and imputation can approximate WGS therefore depends on reference panel, genotype array, sample ancestry, and genomic location. Imputation quality by variant or genomic region can be queried with our new tool, RsqBrowser, now deployed on the Michigan Imputation Server.
Collapse
Affiliation(s)
- Sarah C. Hanks
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Lukas Forer
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sebastian Schönherr
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jonathon LeFaive
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Taylor Martins
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Ryan Welch
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Sarah A. Gagliano Taliun
- Department of Medicine and Department of Neurosciences, Université de Montréal, Montreal, QC, Canada,Research Centre, Montreal Heart Institute, Montreal, QC, Canada
| | - David Braff
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Jill M. Johnsen
- Research Institute, Bloodworks, Seattle, WA, USA,Department of Medicine, University of Washington, Seattle, WA, USA
| | - Eimear E. Kenny
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA,Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Ruth F.J. Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA,Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Carlos Pato
- Departments of Psychiatry, Rutgers University, Robert Wood Johnson Medical School and New Jersey Medical School, New Brunswick, NJ, USA
| | - Michele T. Pato
- Departments of Psychiatry, Rutgers University, Robert Wood Johnson Medical School and New Jersey Medical School, New Brunswick, NJ, USA
| | - Albert V. Smith
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | | | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Laura J. Scott
- Department of Biostatistics and Center for Statistical Genetics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Christian Fuchsberger
- Institute for Biomedicine (Affiliated with the University of Lübeck), Eurac Research, Bolzano, Italy.
| |
Collapse
|
35
|
Mathur R, Fang F, Gaddis N, Hancock DB, Cho MH, Hokanson JE, Bierut LJ, Lutz SM, Young K, Smith AV, Silverman EK, Page GP, Johnson EO. GAWMerge expands GWAS sample size and diversity by combining array-based genotyping and whole-genome sequencing. Commun Biol 2022; 5:806. [PMID: 35953715 PMCID: PMC9372058 DOI: 10.1038/s42003-022-03738-6] [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: 12/21/2021] [Accepted: 07/18/2022] [Indexed: 11/09/2022] Open
Abstract
Genome-wide association studies (GWAS) have made impactful discoveries for complex diseases, often by amassing very large sample sizes. Yet, GWAS of many diseases remain underpowered, especially for non-European ancestries. One cost-effective approach to increase sample size is to combine existing cohorts, which may have limited sample size or be case-only, with public controls, but this approach is limited by the need for a large overlap in variants across genotyping arrays and the scarcity of non-European controls. We developed and validated a protocol, Genotyping Array-WGS Merge (GAWMerge), for combining genotypes from arrays and whole-genome sequencing, ensuring complete variant overlap, and allowing for diverse samples like Trans-Omics for Precision Medicine to be used. Our protocol involves phasing, imputation, and filtering. We illustrated its ability to control technology driven artifacts and type-I error, as well as recover known disease-associated signals across technologies, independent datasets, and ancestries in smoking-related cohorts. GAWMerge enables genetic studies to leverage existing cohorts to validly increase sample size and enhance discovery for understudied traits and ancestries.
Collapse
Affiliation(s)
- Ravi Mathur
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Fang Fang
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Nathan Gaddis
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Dana B Hancock
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
| | - Michael H Cho
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - John E Hokanson
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | - Laura J Bierut
- Department of Psychiatry, Washington University, St. Louis, MO, USA
| | - Sharon M Lutz
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care, Boston, MA, USA
| | - Kendra Young
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA
| | - Albert V Smith
- Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Edwin K Silverman
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Grier P Page
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA
- Fellow Program, RTI International, Research Triangle Park, NC, USA
| | - Eric O Johnson
- GenOmics, Bioinformatics, and Translational Research Center, RTI International, Research Triangle Park, NC, USA.
- Fellow Program, RTI International, Research Triangle Park, NC, USA.
| |
Collapse
|
36
|
Keur N, Ricaño-Ponce I, Kumar V, Matzaraki V. A systematic review of analytical methods used in genetic association analysis of the X-chromosome. Brief Bioinform 2022; 23:6651325. [PMID: 35901513 DOI: 10.1093/bib/bbac287] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/07/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Genetic association studies have been very successful at elucidating the genetic background of many complex diseases/traits. However, the X-chromosome is often neglected in these studies because of technical difficulties and the fact that most tools only utilize genetic data from autosomes. In this review, we aim to provide an overview of different practical approaches that are followed to incorporate the X-chromosome in association analysis, such as Genome-Wide Association Studies and Expression Quantitative Trait Loci Analysis. In general, the choice of which test statistics is most appropriate will depend on three main criteria: (1) the underlying X-inactivation model, (2) if Hardy-Weinberg equilibrium holds and sex-specific allele frequencies are expected and (3) whether adjustment for confounding variables is required. All in all, it is recommended that a combination of different association tests should be used for the analysis of X-chromosome.
Collapse
Affiliation(s)
- Nick Keur
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 HP, Nijmegen, The Netherlands
| | - Isis Ricaño-Ponce
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 HP, Nijmegen, The Netherlands
| | - Vinod Kumar
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 HP, Nijmegen, The Netherlands.,Department of Genetics, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9700RB, Groningen, The Netherlands
| | - Vasiliki Matzaraki
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 HP, Nijmegen, The Netherlands
| |
Collapse
|
37
|
Thanh Nguyen D, Hoang Nguyen Q, Thuy Duong N, Vo NS. LmTag: functional-enrichment and imputation-aware tag SNP selection for population-specific genotyping arrays. Brief Bioinform 2022; 23:6627269. [PMID: 35780383 DOI: 10.1093/bib/bbac252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/02/2022] [Accepted: 05/31/2022] [Indexed: 12/16/2022] Open
Abstract
Despite the rapid development of sequencing technology, single-nucleotide polymorphism (SNP) arrays are still the most cost-effective genotyping solutions for large-scale genomic research and applications. Recent years have witnessed the rapid development of numerous genotyping platforms of different sizes and designs, but population-specific platforms are still lacking, especially for those in developing countries. SNP arrays designed for these countries should be cost-effective (small size), yet incorporate key information needed to associate genotypes with traits. A key design principle for most current platforms is to improve genome-wide imputation so that more SNPs not included in the array (imputed SNPs) can be predicted. However, current tag SNP selection methods mostly focus on imputation accuracy and coverage, but not the functional content of the array. It is those functional SNPs that are most likely associated with traits. Here, we propose LmTag, a novel method for tag SNP selection that not only improves imputation performance but also prioritizes highly functional SNP markers. We apply LmTag on a wide range of populations using both public and in-house whole-genome sequencing databases. Our results show that LmTag improved both functional marker prioritization and genome-wide imputation accuracy compared to existing methods. This novel approach could contribute to the next generation genotyping arrays that provide excellent imputation capability as well as facilitate array-based functional genetic studies. Such arrays are particularly suitable for under-represented populations in developing countries or non-model species, where little genomics data are available while investment in genome sequencing or high-density SNP arrays is limited. $\textrm{LmTag}$ is available at: https://github.com/datngu/LmTag.
Collapse
Affiliation(s)
- Dat Thanh Nguyen
- Center for Biomedical Informatics, Vingroup Big Data Institute, 458 Minh Khai, 10000, Hanoi, Vietnam
| | - Quan Hoang Nguyen
- Institute for Molecular Bioscience, University of Queensland, st Lucia, QLD 4067, Brisbane, Australia
| | - Nguyen Thuy Duong
- Center for Biomedical Informatics, Vingroup Big Data Institute, 458 Minh Khai, 10000, Hanoi, Vietnam.,Institute of Genome Research, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet, 10000, Hanoi, Vietnam
| | - Nam S Vo
- Center for Biomedical Informatics, Vingroup Big Data Institute, 458 Minh Khai, 10000, Hanoi, Vietnam.,College of Engineering and Computer Science, VinUniversity, Vinhomes Ocean Park, 10000, Hanoi, Vietnam
| |
Collapse
|
38
|
Temba GS, Vadaq N, Wan J, Kullaya V, Huskens D, Pecht T, Jaeger M, Boahen CK, Matzaraki V, Broeders W, Joosten LAB, Faradz SMH, Kibiki G, Middeldorp S, Cavalieri D, Lionetti P, de Groot PG, Schultze JL, Netea MG, Kumar V, de Laat B, Mmbaga BT, van der Ven AJ, Roest M, de Mast Q. Differences in thrombin and plasmin generation potential between East African and Western European adults: The role of genetic and non-genetic factors. J Thromb Haemost 2022; 20:1089-1105. [PMID: 35102686 PMCID: PMC9305795 DOI: 10.1111/jth.15657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Geographic variability in coagulation across populations and their determinants are poorly understood. OBJECTIVE To compare thrombin (TG) and plasmin (PG) generation parameters between healthy Tanzanian and Dutch individuals, and to study associations with inflammation and different genetic, host and environmental factors. METHODS TG and PG parameters were measured in 313 Tanzanians of African descent living in Tanzania and 392 Dutch of European descent living in the Netherlands and related to results of a dietary questionnaire, circulating inflammatory markers, genotyping, and plasma metabolomics. RESULTS Tanzanians exhibited an enhanced TG and PG capacity, compared to Dutch participants. A higher proportion of Tanzanians had a TG value in the upper quartile with a PG value in the lower/middle quartile, suggesting a relative pro-coagulant state. Tanzanians also displayed an increased normalized thrombomodulin sensitivity ratio, suggesting reduced sensitivity to protein C. In Tanzanians, PG parameters (lag time and TTP) were associated with seasonality and food-derived plasma metabolites. The Tanzanians had higher concentrations of pro-inflammatory cytokines, which correlated strongly with TG and PG parameters. There was limited overlap in genetic variation associated with TG and PG parameters between the two cohorts. Pathway analysis of genetic variants in the Tanzanian cohort revealed multiple immune pathways that were enriched with TG and PG traits, confirming the importance of co-regulation between coagulation and inflammation. CONCLUSIONS Tanzanians have an enhanced TG and PG potential compared to Dutch individuals, which may relate to differences in inflammation, genetics and diet. These observations highlight the importance of better understanding of the geographic variability in coagulation across populations.
Collapse
Affiliation(s)
- Godfrey S. Temba
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
- Department of Medical Biochemistry and Molecular BiologyKilimanjaro Christian Medical University College (KCMUCo)MoshiTanzania
| | - Nadira Vadaq
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
- Center for Tropical and Infectious Diseases (CENTRID)Faculty of MedicineDr. Kariadi HospitalDiponegoro UniversitySemarangIndonesia
| | - Jun Wan
- Synapse Research InstituteCardiovascular Research Institute MaastrichtMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Vesla Kullaya
- Department of Medical Biochemistry and Molecular BiologyKilimanjaro Christian Medical University College (KCMUCo)MoshiTanzania
- Kilimanjaro Clinical Research InstituteKilimanjaro Christian Medical CenterMoshiTanzania
| | - Dana Huskens
- Synapse Research InstituteCardiovascular Research Institute MaastrichtMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Tal Pecht
- Department for Genomics and ImmunoregulationLife & Medical Sciences (LIMES) InstituteUniversity of BonnBonnGermany
- Systems MedicineGerman Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Martin Jaeger
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
| | - Collins K. Boahen
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
| | - Vasiliki Matzaraki
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
| | - Wieteke Broeders
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
| | - Leo A. B. Joosten
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
| | - Sultana M. H. Faradz
- Division of Human GeneticsCenter for Biomedical Research (CEBIOR)Faculty of MedicineDiponegoro University/Diponegoro National HospitalSemarangIndonesia
| | - Gibson Kibiki
- Kilimanjaro Clinical Research InstituteKilimanjaro Christian Medical CenterMoshiTanzania
| | - Saskia Middeldorp
- Department of Internal MedicineRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
| | | | - Paolo Lionetti
- Departement NEUROFARBAMeyer Children's HospitalUniversity of Florence – Gastroenterology and Nutrition UnitFlorenceItaly
| | - Philip G. de Groot
- Synapse Research InstituteCardiovascular Research Institute MaastrichtMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Joachim L. Schultze
- Department for Genomics and ImmunoregulationLife & Medical Sciences (LIMES) InstituteUniversity of BonnBonnGermany
- Systems MedicineGerman Center for Neurodegenerative Diseases (DZNE)BonnGermany
- PRECISE Platform for Single Cell Genomics and EpigenomicsGerman Center for Neurodegenerative Diseases (DZNE) and University of BonnBonnGermany
| | - Mihai G. Netea
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
- Department for Immunology and MetabolismLife & Medical Sciences (LIMES) InstituteUniversity of BonnBonnGermany
| | - Vinod Kumar
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
- Department of GeneticsUniversity Medical Centre GroningenUniversity of GroningenGroningenthe Netherlands
| | - Bas de Laat
- Synapse Research InstituteCardiovascular Research Institute MaastrichtMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Blandina T. Mmbaga
- Kilimanjaro Clinical Research InstituteKilimanjaro Christian Medical CenterMoshiTanzania
- Department of PaediatricsKilimanjaro Christian Medical University College (KCMUCo)MoshiTanzania
| | - Andre J. van der Ven
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
| | - Mark Roest
- Synapse Research InstituteCardiovascular Research Institute MaastrichtMaastricht University Medical CenterMaastrichtthe Netherlands
| | - Quirijn de Mast
- Department of Internal MedicineRadboudumc Center for Infectious DiseasesRadboud Institute of Health Science (RIHS)Radboud university medical centerNijmegenthe Netherlands
| |
Collapse
|
39
|
Auwerx C, Sadler MC, Reymond A, Kutalik Z. From pharmacogenetics to pharmaco-omics: Milestones and future directions. HGG ADVANCES 2022; 3:100100. [PMID: 35373152 PMCID: PMC8971318 DOI: 10.1016/j.xhgg.2022.100100] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The origins of pharmacogenetics date back to the 1950s, when it was established that inter-individual differences in drug response are partially determined by genetic factors. Since then, pharmacogenetics has grown into its own field, motivated by the translation of identified gene-drug interactions into therapeutic applications. Despite numerous challenges ahead, our understanding of the human pharmacogenetic landscape has greatly improved thanks to the integration of tools originating from disciplines as diverse as biochemistry, molecular biology, statistics, and computer sciences. In this review, we discuss past, present, and future developments of pharmacogenetics methodology, focusing on three milestones: how early research established the genetic basis of drug responses, how technological progress made it possible to assess the full extent of pharmacological variants, and how multi-dimensional omics datasets can improve the identification, functional validation, and mechanistic understanding of the interplay between genes and drugs. We outline novel strategies to repurpose and integrate molecular and clinical data originating from biobanks to gain insights analogous to those obtained from randomized controlled trials. Emphasizing the importance of increased diversity, we envision future directions for the field that should pave the way to the clinical implementation of pharmacogenetics.
Collapse
Affiliation(s)
- Chiara Auwerx
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Marie C. Sadler
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, Lausanne, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Zoltán Kutalik
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- University Center for Primary Care and Public Health, Lausanne, Switzerland
| |
Collapse
|
40
|
Tilch E, Schormair B, Zhao C, Högl B, Stefani A, Berger K, Trenkwalder C, Bachmann CG, Hornyak M, Fietze I, Müller-Nurasyid M, Peters A, Herms S, Nöthen MM, Müller-Myhsok B, Oexle K, Winkelmann J. Exomechip-based rare variant association study in restless legs syndrome. Sleep Med 2022; 94:26-30. [DOI: 10.1016/j.sleep.2022.04.001] [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: 02/09/2022] [Revised: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022]
|
41
|
Mizrahi-Man O, Woehrmann MH, Webster TA, Gollub J, Bivol A, Keeble SM, Aull KH, Mittal A, Roter AH, Wong BA, Schmidt JP. Novel genotyping algorithms for rare variants significantly improve the accuracy of Applied Biosystems™ Axiom™ array genotyping calls: Retrospective evaluation of UK Biobank array data. PLoS One 2022; 17:e0277680. [PMID: 36395175 PMCID: PMC9671364 DOI: 10.1371/journal.pone.0277680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 11/01/2022] [Indexed: 11/18/2022] Open
Abstract
The UK Biobank genotyped about 500k participants using Applied Biosystems Axiom microarrays. Participants were subsequently sequenced by the UK Biobank Exome Sequencing Consortium. Axiom genotyping was highly accurate in comparison to sequencing results, for almost 100,000 variants both directly genotyped on the UK Biobank Axiom array and via whole exome sequencing. However, in a study using the exome sequencing results of the first 50k individuals as reference (truth), it was observed that the positive predictive value (PPV) decreased along with the number of heterozygous array calls per variant. We developed a novel addition to the genotyping algorithm, Rare Heterozygous Adjusted (RHA), to significantly improve PPV in variants with minor allele frequency below 0.01%. The improvement in PPV was roughly equal when comparing to the exome sequencing of 50k individuals, or to the more recent ~200k individuals. Sensitivity was higher in the 200k data. The improved calling algorithm, along with enhanced quality control of array probesets, significantly improved the positive predictive value and the sensitivity of array data, making it suitable for the detection of ultra-rare variants.
Collapse
Affiliation(s)
- Orna Mizrahi-Man
- Thermo Fisher Scientific, Santa Clara, CA, United States of America
| | | | | | - Jeremy Gollub
- Thermo Fisher Scientific, Santa Clara, CA, United States of America
| | - Adrian Bivol
- Thermo Fisher Scientific, Santa Clara, CA, United States of America
| | - Sara M. Keeble
- Thermo Fisher Scientific, Santa Clara, CA, United States of America
| | | | - Anuradha Mittal
- Thermo Fisher Scientific, Santa Clara, CA, United States of America
| | - Alan H. Roter
- Thermo Fisher Scientific, Santa Clara, CA, United States of America
| | - Brant A. Wong
- Thermo Fisher Scientific, Santa Clara, CA, United States of America
| | - Jeanette P. Schmidt
- Thermo Fisher Scientific, Santa Clara, CA, United States of America
- * E-mail:
| |
Collapse
|
42
|
Neumann GB, Korkuć P, Arends D, Wolf MJ, May K, Reißmann M, Elzaki S, König S, Brockmann GA. Design and performance of a bovine 200 k SNP chip developed for endangered German Black Pied cattle (DSN). BMC Genomics 2021; 22:905. [PMID: 34922441 PMCID: PMC8684242 DOI: 10.1186/s12864-021-08237-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 12/03/2021] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND German Black Pied cattle (DSN) are an endangered dual-purpose breed which was largely replaced by Holstein cattle due to their lower milk yield. DSN cattle are kept as a genetic reserve with a current herd size of around 2500 animals. The ability to track sequence variants specific to DSN could help to support the conservation of DSN's genetic diversity and to provide avenues for genetic improvement. RESULTS Whole-genome sequencing data of 304 DSN cattle were used to design a customized DSN200k SNP chip harboring 182,154 variants (173,569 SNPs and 8585 indels) based on ten selection categories. We included variants of interest to DSN such as DSN unique variants and variants from previous association studies in DSN, but also variants of general interest such as variants with predicted consequences of high, moderate, or low impact on the transcripts and SNPs from the Illumina BovineSNP50 BeadChip. Further, the selection of variants based on haplotype blocks ensured that the whole-genome was uniformly covered with an average variant distance of 14.4 kb on autosomes. Using 300 DSN and 162 animals from other cattle breeds including Holstein, endangered local cattle populations, and also a Bos indicus breed, performance of the SNP chip was evaluated. Altogether, 171,978 (94.31%) of the variants were successfully called in at least one of the analyzed breeds. In DSN, the number of successfully called variants was 166,563 (91.44%) while 156,684 (86.02%) were segregating at a minor allele frequency > 1%. The concordance rate between technical replicates was 99.83 ± 0.19%. CONCLUSION The DSN200k SNP chip was proved useful for DSN and other Bos taurus as well as one Bos indicus breed. It is suitable for genetic diversity management and marker-assisted selection of DSN animals. Moreover, variants that were segregating in other breeds can be used for the design of breed-specific customized SNP chips. This will be of great value in the application of conservation programs for endangered local populations in the future.
Collapse
Affiliation(s)
- Guilherme B Neumann
- Animal Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer-Institute for Agricultural and Horticultural Sciences, Humboldt Universität zu Berlin, Berlin, Germany
| | - Paula Korkuć
- Animal Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer-Institute for Agricultural and Horticultural Sciences, Humboldt Universität zu Berlin, Berlin, Germany
| | - Danny Arends
- Animal Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer-Institute for Agricultural and Horticultural Sciences, Humboldt Universität zu Berlin, Berlin, Germany
| | - Manuel J Wolf
- Institute of Animal Breeding and Genetics, Justus-Liebig-Universität, Gießen, Germany
| | - Katharina May
- Institute of Animal Breeding and Genetics, Justus-Liebig-Universität, Gießen, Germany
| | - Monika Reißmann
- Animal Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer-Institute for Agricultural and Horticultural Sciences, Humboldt Universität zu Berlin, Berlin, Germany
| | - Salma Elzaki
- Animal Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer-Institute for Agricultural and Horticultural Sciences, Humboldt Universität zu Berlin, Berlin, Germany.,Department of Genetics and Animal Breeding, Faculty of Animal Production, University of Khartoum, Khartoum North, Sudan
| | - Sven König
- Institute of Animal Breeding and Genetics, Justus-Liebig-Universität, Gießen, Germany
| | - Gudrun A Brockmann
- Animal Breeding Biology and Molecular Genetics, Albrecht Daniel Thaer-Institute for Agricultural and Horticultural Sciences, Humboldt Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
43
|
Genotyping arrays, population genetic studies and clinical implications. Eur J Hum Genet 2021; 29:1591-1592. [PMID: 34616014 DOI: 10.1038/s41431-021-00979-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|