1
|
Avila Martins CC, Maschietto M, Kimura L, Alvizi L, Nunes K, Magalhães Borges V, Victorino Krepischi AC, Mingroni-Netto RC. Differential methylation in blood pressure control genes is associated to essential hypertension in African Brazilian populations. Epigenetics 2025; 20:2477850. [PMID: 40143670 PMCID: PMC11951699 DOI: 10.1080/15592294.2025.2477850] [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: 11/27/2024] [Revised: 02/23/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
While genetic studies have provided insights into essential hypertension (EH, defined by high blood pressure ≥140/90 mmHg), investigation through epigenetics may address gaps in understanding its heritability. This study focused on African Brazilian populations in Vale do Ribeira River region, due to their high hypertension prevalence. We aimed to determine if DNA methylation is linked to hypertension susceptibility, through a genome-wide evaluation of 80 peripheral blood samples from normotensive (39) and hypertensive (41) individuals, with Infinium Methylation EPIC BeadChip platform. Data were analyzed using ChAMP package and cross-referenced with information from databases such as EWAS Atlas, GWAS catalog, GeneCards, literature, and tools such as VarElect and EWAS Toolkit. The comparison between hypertensive and normotensive revealed 190 differentially methylated CpG positions (DMPs) and 46 differentially methylated regions (DMRs), both with p-value ≤0.05. Among the DMPs, 27 were found to have a plausible role in blood pressure. Among the DMRs, those mapped to ABAT, BLCAP, CERS3, EIF4E, FMN1, GABBR1, HLA-DQB2, HOXA5, IL5RA, KCNH2, MIR487B, MIR539, MIR886, MKRN3, NUDT12, PON3, RNF39, RWDD3, and TSHBS1 were highlighted because of their lowest p-values, current literature, and/or VarElect prioritization. Our findings suggest that differences in methylation contribute to the high susceptibility to essential hypertension in these populations.
Collapse
Affiliation(s)
- Camila Cristina Avila Martins
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | | | - Lilian Kimura
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lucas Alvizi
- Cell & Development Biology, University College London, London, UK
| | - Kelly Nunes
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Vinícius Magalhães Borges
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, USA
| | - Ana Cristina Victorino Krepischi
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Regina Célia Mingroni-Netto
- Centro de Estudos sobre o Genoma Humano e Células Tronco, Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
2
|
Wang YX, Fei CJ, Shen C, Ou YN, Liu WS, Yang L, Wu BS, Deng YT, Feng JF, Cheng W, Yu JT. Exome sequencing identifies protein-coding variants associated with loneliness and social isolation. J Affect Disord 2025; 375:192-204. [PMID: 39842675 DOI: 10.1016/j.jad.2025.01.096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 10/31/2024] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Loneliness and social isolation are serious yet underappreciated public health problems, with their genetic underpinnings remaining largely unknown. We aimed to explore the role of protein-coding variants in the manifestation of loneliness and social isolation. METHODS We conducted the first exome-wide association analysis on loneliness and social isolation, utilizing 336,115 participants of white-British ancestry for loneliness and 346,115 for social isolation. Sensitivity analyses were performed to validate the genetic findings. We estimated the genetic burden heritability of loneliness and social isolation and provided biological insights into them. RESULTS We identified six novel risk genes (ANKRD12, RIPOR2, PTEN, ARL8B, NF1, and PIMREG) associated with loneliness and two (EDARADD and GIGYF1) with social isolation through analysis of rare coding variants. Brain-wide association analysis uncovered 47 associations between identified genes and brain structure phenotypes, many of which are critical for social processing and interaction. Phenome-wide association analysis established significant links between these genes and phenotypes across five categories, mostly blood biomarkers and cognitive measures. LIMITATIONS The measurements of loneliness and social isolation in UK Biobank are brief for these multi-layer social factors, some relevant aspects may be missed. CONCLUSIONS Our study revealed 13 risk genes associated with loneliness and 6 with social isolation, with the majority being novel discoveries. These findings advance our understanding of the genetic basis of these two traits. The study provides a foundation for future studies aimed at exploring the functional mechanisms of these genes and their potential implications for public health interventions targeting loneliness and social isolation.
Collapse
Affiliation(s)
- Yi-Xuan Wang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Chen-Jie Fei
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Chun Shen
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China; Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai, China; Department of Computer Science, University of Warwick, Coventry CV4 7AL, UK.
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
3
|
Dommisch H, Hoedke D, Lu EMC, Schäfer A, Richter G, Kang J, Nibali L. Genetic Biomarkers for Periodontal Diseases: A Systematic Review. J Clin Periodontol 2025. [PMID: 40197750 DOI: 10.1111/jcpe.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 04/10/2025]
Abstract
AIMS To identify genetic biomarkers that may be used in the diagnosis, prevention or management of different forms of periodontal disease. MATERIALS AND METHODS Following protocol registration and PICOTS (patient, intervention, comparison, outcome, time, studies) questions, a systematic search of the literature was conducted (PudMed, Ovid), resulting in 1592 papers screened by two reviewers. Diagnostic data were extracted or calculated from included papers and compared with clinically determined diagnoses, disease progression and/or response to treatment. RESULTS A total of 607 articles met the inclusion criteria, including 10 reporting data on gingivitis and 597 on periodontitis. Only two papers reported diagnostic performance data, while for 41 articles on large candidate gene studies, diagnostic performance could be calculated from the reported data. No study using chair-side tests was identified. Low to moderate values for sensitivity, specificity, positive and negative predictive value and diagnostic accuracy were found. CONCLUSION No genetic diagnostic test of clinical value emerged for periodontal diagnosis, prevention or prediction of disease resolution. Thus, potential future applications of polygenic risk scores that encode susceptibility, as well as single-marker testing for monogenic or oligogenic forms of periodontal diseases, are discussed.
Collapse
Affiliation(s)
- H Dommisch
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - D Hoedke
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - E M-C Lu
- Periodontology Unit, Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - A Schäfer
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - G Richter
- Department of Periodontology, Oral Medicine and Oral Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - J Kang
- Periodontology Unit, Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - L Nibali
- Periodontology Unit, Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| |
Collapse
|
4
|
Hashiba T, Sugawara Y, Hirakawa Y, Sato D, Inagi R, Nangaku M. Pathogenic variants prevalence patients with diabetic kidney disease in Japan: A descriptive study. J Diabetes Investig 2025. [PMID: 40197820 DOI: 10.1111/jdi.70041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/10/2025] Open
Abstract
AIMS/INTRODUCTION The impact of rare pathogenic variants on diabetic kidney disease (DKD) has not been investigated in detail. Previous studies have detected pathogenic variants in 22% of Caucasian patients with DKD; however, this proportion may vary depending on ethnicity and updates to the database. Therefore, we performed a whole-genome analysis of patients with DKD in type 2 diabetes mellitus in Japan, utilizing a recent database to investigate the prevalence of kidney-related pathogenic variants and describe the characteristics of these patients. MATERIALS AND METHODS Whole-genome sequencing was performed, and variants were analyzed following the GATK Best Practices. We extracted data on 790 genes associated with Mendelian kidney and genitourinary diseases. Pathogenic variants were defined based on the American College of Medical Genetics criteria, including both heterozygous and homozygous variants classified as pathogenic or likely pathogenic. RESULTS Among 79 participants, heterozygous pathogenic variants were identified in 27 (34.1%), a higher prevalence than previously reported. No homozygous pathogenic variants were detected. The identified heterozygous pathogenic variants were roughly divided into 23.7% related to glomerulopathy, 36.8% related to tubulointerstitial disease, 10.5% related to cystic disease/ciliopathy, and 28.9% related to others. Diagnostic variants were found in 10 patients (12.7%) in seven genes (ABCC6, ALPL, ASXL1, BMPR2, GCM2, PAX2, and WT1), all associated with autosomal dominant congenital disease. CONCLUSIONS This study identified a considerable number of patients with DKD in Japan who carried kidney-related heterozygous pathogenic variants. These findings suggest potential ethnic differences and highlight the impact of database updates on variant detection.
Collapse
Affiliation(s)
- Toyohiro Hashiba
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuka Sugawara
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yosuke Hirakawa
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Dai Sato
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Reiko Inagi
- Division of Chronic Kidney Disease Pathophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masaomi Nangaku
- Division of Nephrology and Endocrinology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
5
|
Faisal F, Danelakis A, Bjørk MH, Winsvold B, Matharu M, Nachev P, Hagen K, Tronvik E, Stubberud A. Prediction of new-onset migraine using clinical-genotypic data from the HUNT Study: a machine learning analysis. J Headache Pain 2025; 26:70. [PMID: 40197205 DOI: 10.1186/s10194-025-02014-2] [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: 03/02/2025] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Migraine is associated with a range of symptoms and comorbid disorders and has a strong genetic basis, but the currently identified risk loci only explain a small portion of the heritability, often termed the "missing heritability". We aimed to investigate if machine learning could exploit the combination of genetic data and general clinical features to identify individuals at risk for new-onset migraine. METHOD This study was a population-based cohort study of adults from the second and third Trøndelag Health Study (HUNT2 and HUNT3). Migraine was captured in a validated questionnaire and based on modified criteria of the International Classification of Headache Disorders (ICHD) and participants underwent genome-wide genotyping. The primary outcome was new-onset migraine defined as a change in disease status from headache-free in HUNT2 to migraine in HUNT3. The migraine risk variants identified in the largest GWAS meta-analysis of migraine were used to identify genetic input features for the models. The general clinical features included demographics, selected comorbidities, medication and stimulant use and non-headache symptoms as predictive factors. Several standard machine learning architectures were constructed, trained, optimized and scored with area under the receiver operating characteristics curve (AUC). The best model during training and validation was used on unseen test sets. Different methods for model explainability were employed. RESULTS A total of 12,995 individuals were included in the predictive modelling (491 new-onset cases). A total of 108 genetic variants and 67 general clinical variables were included in the models. The top performing decision-tree classifier achieved a test set AUC of 0.56 when using only genotypic data, 0.68 when using only clinical data and 0.72 when using both genetic and clinical data. Combining the genotype only and clinical data only models resulted in a lower predictivity with an AUC of 0.67. The most important clinical features were age, marital status and work situation as well as several genetic variants. CONCLUSION The combination of genotype and routine demographic and non-headache clinical data correctly predict the new onset of migraine in approximately 2 out of 3 cases, supporting that there are important genotypic-phenotypic interactions partaking in the new-onset of migraine.
Collapse
Affiliation(s)
- Fahim Faisal
- Norhead Norwegian Centre for Headache Research, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neuromedicine and Movement Science, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Antonios Danelakis
- Norhead Norwegian Centre for Headache Research, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- Department of Computer Science, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Marte-Helene Bjørk
- Norhead Norwegian Centre for Headache Research, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Bendik Winsvold
- Norhead Norwegian Centre for Headache Research, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- Department of Research and Innovation, Division of Clinical Neuroscience, Oslo University Hospital, Oslo, Norway
- Department of Neurology, Oslo University Hospital, Oslo, Norway
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Manjit Matharu
- Norhead Norwegian Centre for Headache Research, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Parashkev Nachev
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Knut Hagen
- Norhead Norwegian Centre for Headache Research, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- Neuroclinic, St Olav University Hospital, Trondheim, Norway
| | - Erling Tronvik
- Norhead Norwegian Centre for Headache Research, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- Department of Neuromedicine and Movement Science, NTNU Norwegian University of Science and Technology, Trondheim, Norway
- Neuroclinic, St Olav University Hospital, Trondheim, Norway
| | - Anker Stubberud
- Norhead Norwegian Centre for Headache Research, NTNU Norwegian University of Science and Technology, Trondheim, Norway.
- Department of Neuromedicine and Movement Science, NTNU Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
6
|
Prasad RB, Hakaste L, Tuomi T. Clinical use of polygenic scores in type 2 diabetes: challenges and possibilities. Diabetologia 2025:10.1007/s00125-025-06419-1. [PMID: 40186687 DOI: 10.1007/s00125-025-06419-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/30/2025] [Indexed: 04/07/2025]
Abstract
Resulting from a combination of genetic and environmental factors, type 2 diabetes is highly heterogeneous in manifestation and disease progression, with the only common feature being chronic hyperglycaemia. In spite of vigorous efforts to elucidate the pathogenetic origins and natural course of the disease, there is still a lack of biomarkers and tools for prevention, disease stratification and treatment. Genome-wide association studies have reported over 1200 variants associated with type 2 diabetes, and the decreased cost of generating genetic data has facilitated the development of polygenic scores for estimating an individual's genetic disease risk based on combining effects from most-or all-genetic variants. In this review, we summarise the current knowledge on type 2 diabetes-related polygenic scores in different ancestries and outline their possible clinical role. We explore the potential applicability of type 2 diabetes polygenic scores to quantify genetic liability for prediction, screening and risk stratification. Given that most genetic risk loci are determined from populations of European origin while other ancestries are under-represented, we also discuss the challenges around their global applicability. To date, the potential for clinical utility of polygenic scores for type 2 diabetes is limited, with such scores outperformed by clinical measures. In the future, rather than predicting risk of type 2 diabetes, the value of polygenic scores may be in stratification of the severity of disease (risk for comorbidities) and treatment response, in addition to aiding in dissecting the pathophysiological mechanisms involved.
Collapse
Affiliation(s)
- Rashmi B Prasad
- Lund University Diabetes Centre, Department of Clinical Sciences, Genetics and Diabetes, CRC, Lund University, Malmö, Sweden.
- Institute for Molecular Medicine Finland (FIMM), Helsinki University, Helsinki, Finland.
| | - Liisa Hakaste
- Institute for Molecular Medicine Finland (FIMM), Helsinki University, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
| | - Tiinamaija Tuomi
- Lund University Diabetes Centre, Department of Clinical Sciences, Genetics and Diabetes, CRC, Lund University, Malmö, Sweden
- Institute for Molecular Medicine Finland (FIMM), Helsinki University, Helsinki, Finland
- Folkhalsan Research Centre, Helsinki, Finland
- Helsinki University Hospital, Abdominal Centre/Endocrinology, Helsinki, Finland
| |
Collapse
|
7
|
Sukhija N, Kanaka KK, Ganguly I, Dixit S, Singh S, Goli RC, Rathi P, Nandini PB, Koloi S. Cataloging copy number variation regions and allied diversity in goat breeds spanning pan India. Mamm Genome 2025:10.1007/s00335-025-10122-2. [PMID: 40175574 DOI: 10.1007/s00335-025-10122-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 03/07/2025] [Indexed: 04/04/2025]
Abstract
Huge genetic diversity is evident among the diverse goat breeds in terms of production, reproduction, adaptability, growth, disease resistance and thermo-tolerance. This diversity is an outcome of both natural and artificial selection acting on the caprine genome over the years. A fine characterization of whole genome variation is now possible by employing Next Generation Sequencing (NGS) technologies. To explore underlying genetics, genome-wide analysis of genetic markers is the best resolution. The study strived to capture variation in terms of CNV/CNVRs among 11 Indian goat breeds. In this study, the first ever resequencing-based CNV/CNVR distribution of Indigenous goat breeds was delineated, providing a sizable addition to the prior caprine CNVRs reported. Different diversity metrics were analyzed using identified CNVR. Principal component analysis (PCA) showed separate clustering of Kanniadu (KAN) and Jharkhand Black (JB) from other breeds under the study, indicating their unique genetic profile as the former breeds were sampled from institutional farms. The admixture analysis and introgression revealed by f3 statistics suggested distinct genetic structuring of JB, KAN and TEL(Tellicherry) as compared to the rest of the studied populations. Apart from this, we also identified 32 selection signatures through VST (Variance-stabilizing transformation) method and key genes such as ZBTB7C, BHLHE22, AGT were found elucidating the genetic architecture of hot and cold adaptation in Indian goats. Information generated hereby in the form of 32,711 autosomal CNVRs and the custom scripts ( https://github.com/kkokay07/Climate-Variables-Analysis.git , https://github.com/chau-mau/SelectCNVR.git and https://github.com/chau-mau/CNVrecaller.git ) will be of relevance in further studies on copy number based genetics.
Collapse
Affiliation(s)
- Nidhi Sukhija
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - K K Kanaka
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Indrajit Ganguly
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India.
| | - Satpal Dixit
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
| | - Sanjeev Singh
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
| | - Rangasai Chandra Goli
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Pallavi Rathi
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - P B Nandini
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Subrata Koloi
- ICAR-National Bureau of Animal Genetic Resources, Karnal, Haryana, 132001, India
- ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| |
Collapse
|
8
|
Lafci NG, Yilmaz B, Yildiz BO. PCOS - the many faces of a disorder in women and men. J Endocrinol Invest 2025; 48:785-798. [PMID: 39680364 DOI: 10.1007/s40618-024-02512-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is a very common endocrine, metabolic and reproductive disorder. The underlying pathophysiology is not yet fully understood and both genetic and environmental factors contribute to its development. We aimed to explore clinical and genetic aspects of familial clustering in PCOS, shedding light on its reproductive and metabolic consequences in both male and female first-degree relatives of the affected women. METHODS Searching the electronic database of PubMed up to October 2023, we synthesized findings from available prospective and retrospective studies and review articles, investigating the familial clustering of PCOS and incorporating data on its metabolic consequences and genetic associations. RESULTS There is a significant clustering of reproductive and metabolic abnormalities in first-degree relatives of women with PCOS. Genetic studies, including genome-wide association studies (GWAS), reveal a complex molecular etiology, emphasizing polygenic architecture. This is supported by the identification of two distinct PCOS subtypes, termed "reproductive" and "metabolic" which exhibit differential genetic underpinnings. CONCLUSION Clinicians should be aware of increased reproductive and metabolic dysfunction both in female and male first-degree relatives of PCOS probands. Current challenges include refining genetic risk scores and understanding the impact of PCOS genetic factors on diverse outcomes, necessitating a sex-specific approach in research and clinical practice. Future directions should address causality, improve diagnostic capability, and unravel the long-term consequences in both genders, emphasizing the importance of proactive clinical assessment in PCOS probands and their families.
Collapse
Affiliation(s)
- Naz Guleray Lafci
- Department of Medical Genetics, Hacettepe University School of Medicine, Ankara, Turkey
| | - Bulent Yilmaz
- Department of Obstetrics and Gynecology, Divison of Reproductive Endocrinology and Infertility, Recep Tayyip Erdoğan University School of Medicine, Rize, Turkey
| | - Bulent Okan Yildiz
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Hacettepe University School of Medicine, Ankara, Turkey.
| |
Collapse
|
9
|
Xu L, Feng Z, Dai Z, Qiu Y, Wu Z, Zhu Z. Novel rare variation of CCDC40 plays a role in the development of idiopathic scoliosis possibly via dysfunction of cilia motility. Spine J 2025; 25:797-804. [PMID: 39662682 DOI: 10.1016/j.spinee.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/22/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024]
Abstract
BACKGROUND CONTEXT Motile cilia dysfunction was reported to lead to scoliosis-like phenotypes in zebrafish models. There is still a lack of population-based study supporting the role of cilia motility associated genes in the etiology of idiopathic scoliosis (IS). PURPOSE To investigate the molecular mechanism underlying the relationship between cilia motility associated genes and the development of adolescent idiopathic scoliosis (AIS). STUDY DESIGN Population-based genetic study. METHODS A cohort of 56 female AIS patients and 30 age-matched nonscoliotic controls were included for tissue expression analysis. 28 patients with lower CCDC40 expression were selected for the exon sequencing. The novel variation was replicated in an independent cohort of 1326 AIS patients and 954 healthy controls. Exogenous versions of WT or mutant human CCDC40 mRNAs were expressed in zebrafish and the phenotype of body axis curvature was observed. RESULTS CCDC40 was found significantly down-expressed in AIS patients as compared with the nonscoliotic controls. A novel coding variant rs185157579 (c.1459G>A) was found significantly associated with AIS, with the mutant allele A adding to the risk of AIS by 2.44 folds. Zebrafish embryo injected with CCDC40 mRNAs containing mutant c.1459G>A presented significantly higher incidence of scoliosis-like phenotype than the wild group. CONCLUSIONS The mutation c.1459G>A in the exon 10 of CCDC40 may lead to body axis curvature of zebrafish by impacting mRNA expression. The underlying molecular mechanism is worthy of further investigation. CLINICAL SIGNIFICANCE Our findings shed a new light on the etiopathogenesis of AIS. The downstream signaling of CCDC40 may be candidate for potential drug targets to prevent the development of AIS. Moreover, the novel variation can be used as a genetic marker of polygenic risk score predicting the risk of AIS.
Collapse
Affiliation(s)
- Leilei Xu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhenhua Feng
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhicheng Dai
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yong Qiu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhichong Wu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zezhang Zhu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China.
| |
Collapse
|
10
|
Zhang K, Zhao J, Mi S, Liu J, Luo J, Liu J, Shi H. Whole-genome variants resource of 298 Saanen dairy goats. Sci Data 2025; 12:528. [PMID: 40157930 PMCID: PMC11954942 DOI: 10.1038/s41597-025-04880-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/21/2025] [Indexed: 04/01/2025] Open
Abstract
The Saanen breeds are often used as terminal sires for hybridization and play an important role in the global dairy food industry. However, there is still a lack of genomics information on the Saanen dairy goats. Whole-genome sequencing offers a promising approach to identify genetic markers associated with economic traits and discover new candidate genes. This can effectively utilize genetic resources to accelerate breeding processes and improve lactation performance in Saanen dairy goats. In this study, we present the genomes of 298 Saanen dairy goats. Through rigorous sequencing and quality control, we achieved an average sequencing depth of 14.6X, with 92.3% of high-quality (Q30 > 90%) data and an average mapping ratio of 99.9%, indicating reliable results. By comparing our data to a reference genome of Saanen dairy goats, we identified14.59 million single nucleotide polymorphisms (SNPs) and 1.34 million insertions-deletions (InDels). This dataset significantly contributes to enriching public databases in dairy goats and provides valuable resources for studying genetic diversity, improving breeds, and developing new varieties.
Collapse
Affiliation(s)
- Kai Zhang
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianqing Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Shirong Mi
- Beijing Compass Biotechnology Co., Ltd, Beijing, 102600, China
| | - Jiqiang Liu
- Beijing Compass Biotechnology Co., Ltd, Beijing, 102600, China
| | - Jun Luo
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jianxin Liu
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hengbo Shi
- Institute of Dairy Science, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
- Zhejiang Key Laboratory of Cow Genetic Improvement & Milk Quality Research, Ministry of Education Key Laboratory of Molecular Animal Nutrition, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
11
|
Smeland OB, Busch C, Andreassen OA, Manchia M. Novel multimodal precision medicine approaches and the relevance of developmental trajectories in bipolar disorder. Biol Psychiatry 2025:S0006-3223(25)01098-4. [PMID: 40157588 DOI: 10.1016/j.biopsych.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/14/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
There is a pressing need to establish objective measures to improve diagnosis, prediction, prevention and treatment of bipolar disorder (BD). Multimodal artificial intelligence (AI) tools could provide these means by incorporating various layers of data orthogonally related to BD, including genomics and other omics, environmental exposures, imaging measures, electronic health records, cognition, sensing devices and clinical variables. These rapidly evolving AI models hold promise to capture the multidimensional complexity of BD and delineate clinically relevant developmental trajectories that could guide clinical care and therapeutic strategies. In this review, we describe the potential of mapping developmental trajectories underlying BD, outline how novel multimodal models could improve the prediction of BD and related outcomes, and discuss specific clinical use cases and key ethical and practical challenges regarding the development and potential implementation of these multimodal AI solutions to advance precision medicine approaches in BD.
Collapse
Affiliation(s)
- Olav B Smeland
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Cecilie Busch
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole A Andreassen
- Centre for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental disorders, University of Oslo, Oslo, Norway.
| | - Mirko Manchia
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari; Unit of Clinical Psychiatry, Department of Medicine, University Hospital Agency of Cagliari; Department of Pharmacology, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
12
|
Tonnele H, Chen D, Morillo F, Garcia-Calleja J, Chitre AS, Johnson BB, Sanches TM, Bonder MJ, Gonzalez A, Kosciolek T, George AM, Han W, Holl K, Horvath A, Ishiwari K, King CP, Lamparelli AC, Martin CD, Martinez AG, Netzley AH, Tripi JA, Wang T, Bosch E, Doris PA, Stegle O, Chen H, Flagel SB, Meyer PJ, Richards JB, Robinson TE, Woods LCS, Polesskaya O, Knight R, Palmer AA, Baud A. Novel insights into the genetic architecture and mechanisms of host/microbiome interactions from a multi-cohort analysis of outbred laboratory rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644349. [PMID: 40166210 PMCID: PMC11957159 DOI: 10.1101/2025.03.20.644349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The intestinal microbiome influences health and disease. Its composition is affected by host genetics and environmental exposures. Understanding host genetic effects is critical but challenging in humans, due to the difficulty of detecting, mapping and interpreting them. To address this, we analysed host genetic effects in four cohorts of outbred laboratory rats exposed to distinct but controlled environments. We found that polygenic host genetic effects were consistent across environments. We identified three replicated microbiome-associated loci. One involved a sialyltransferase gene and Paraprevotella and we found a similar association, between ST6GAL1 and Paraprevotella , in a human cohort. Given Paraprevotella 's known immunity-potentiating functions, this suggests ST6GAL1 's effects on IgA nephropathy and COVID-19 breakthrough infections may be mediated by Paraprevotella . Moreover, we found evidence of indirect genetic effects on microbiome phenotypes, which substantially increased their total genetic variance. Finally, we identified a novel mechanism whereby indirect genetic effects can contribute to "missing heritability".
Collapse
Affiliation(s)
- Helene Tonnele
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Denghui Chen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Felipe Morillo
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| | - Jorge Garcia-Calleja
- Institute of Evolutionary Biology (CSIC-UPF), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Apurva S Chitre
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Benjamin B Johnson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | | | - Marc Jan Bonder
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Antonio Gonzalez
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Tomasz Kosciolek
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Anthony M George
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY, USA8
| | - Wenyan Han
- Department of Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Katie Holl
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Aidan Horvath
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Keita Ishiwari
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY, USA8
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | | | | | - Connor D Martin
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY, USA8
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Angel Garcia Martinez
- Department of Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Alesa H Netzley
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Jordan A Tripi
- Department of Psychology, University at Buffalo, NY, USA
| | - Tengfei Wang
- Department of Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Elena Bosch
- Institute of Evolutionary Biology (CSIC-UPF), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Peter A Doris
- Center for Human Genetics, Institute of Molecular Medicine, McGovern Medical School, University of Texas at Houston, TX, USA
| | - Oliver Stegle
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Hao Chen
- Department of Pharmacology, Addiction Science and Toxicology, University of Tennessee Health Sciences Center, Memphis, TN, USA
| | - Shelly B. Flagel
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Paul J Meyer
- Department of Psychology, University at Buffalo, NY, USA
| | - Jerry B Richards
- Clinical and Research Institute on Addictions, University at Buffalo, Buffalo, NY, USA8
- Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, NY, USA
| | - Terry E. Robinson
- Department of Psychology, University of Michigan, Ann Arbor, MI, USA
| | - Leah C Solberg Woods
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest University School of Medicine, Winston Salem, NC, USA
| | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science & Engineering, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, La Jolla, CA, San Diego, USA
| | - Abraham A Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Amelie Baud
- Centre for Genomic Regulation, Barcelona Institute of Science and Technology, Spain
- Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
13
|
Tomasi D, Volkow ND. Brain asymmetry and its association with inattention and heritability during neurodevelopment. Transl Psychiatry 2025; 15:96. [PMID: 40140344 PMCID: PMC11947263 DOI: 10.1038/s41398-025-03327-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/23/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
The relationship between brain asymmetry and inattention, and their heritability is not well understood. Utilizing advanced neuroimaging, we examined brain asymmetry with data from the Adolescent Brain Cognitive Development (ABCD; n = 8943; 9-10 y) and the Human Connectome Project (HCP) cohorts (n = 1033; 5-100 y). Data-driven metrics from resting-state fMRI and morphometrics revealed reproducible and stable brain asymmetry patterns across the lifespan. In children, high levels of inattention were highly heritable (61%) and linked to reduced leftward asymmetry of functional connectivity in the dorsal posterior superior temporal sulcus (dpSTS), a region interconnected with a left-lateralized language network. However, reduced dpSTS asymmetry had low heritability (16%) and was associated with lower cognitive performance suggesting that non-genetic factors, such as those mediating cognitive performance, might underlie its association with dpSTS asymmetry. Interventions that enhance cognition might help optimize brain function and reduce inattention.
Collapse
Affiliation(s)
- Dardo Tomasi
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA.
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, USA
| |
Collapse
|
14
|
Hamazaki K, Iwata H, Mary-Huard T. A novel genome-wide association study method for detecting quantitative trait loci interacting with complex population structures in plant genetics. Genetics 2025:iyaf038. [PMID: 40091626 DOI: 10.1093/genetics/iyaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
In plant genetics, most modern association analyses are performed on panels that bring together individuals from several populations, including admixed individuals whose genomes comprise chromosomal regions from different populations. These panels can identify quantitative trait loci (QTLs) with population-specific effects and epistatic interactions between QTLs and polygenic backgrounds. However, analyzing a diverse panel constitutes a challenge for statistical analysis. The statistical model must account for possible interactions between a QTL and the panel structure while strictly controlling the detection error rate. Although models to detect population-specific QTLs have already been developed, they rely on prior information about the population structure. In practice, this prior information may be missing as many genome-wide association study (GWAS) panels exhibit complex population structures. The present study introduces 2 new models for detecting QTLs interacting with complex population structures. Both incorporate an interaction term between single nucleotide polymorphism/haplotype block and genetic background into conventional GWAS models. The proposed models were compared with state-of-the-art models through simulation studies that considered QTLs with different levels of interaction with their genetic backgrounds. Results showed that models matching simulation settings were most effective for detecting corresponding QTLs while the proposed models outperformed classical models in detecting QTLs interacting with polygenes. Additionally, when applied to a soybean dataset, one of our models identified putative associated QTLs that conventional models failed to detect. The new models, implemented in the RAINBOWR package available on CRAN, are expected to help uncover complex trait genetic architectures.
Collapse
Affiliation(s)
- Kosuke Hamazaki
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Hiroyoshi Iwata
- Department of Agricultural and Environmental Biology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo 113-8657, Japan
| | - Tristan Mary-Huard
- MIA-Paris Saclay, INRAE, AgroParisTech, Université Paris-Saclay, Palaiseau 91120, France
- Université Paris-Saclay, INRAE, CNRS, AgroParisTech, Génétique Quantitative et Evolution-Le Moulon, Gif-sur-Yvette 91190, France
| |
Collapse
|
15
|
Kim YM, Seong HS, Ha SJ, Kim YS, Kim JK, Baek H, Kwon S, Yoon S, Lee JH, Seo D, Chung WH, Hong JK, Choi JW, Cho ES. Detection of Copy Number Variations in Woori-Heukdon Populations with the Illumina PorcineSNP60 Bead-Chip Array. Animals (Basel) 2025; 15:774. [PMID: 40150303 PMCID: PMC11939295 DOI: 10.3390/ani15060774] [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: 12/23/2024] [Revised: 02/22/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
This study investigated copy number variations (CNVs) in 2112 pigs from five populations: Korean Duroc (DUC), Korean Native Pig (KNP), and their crossbred offspring (F1, F2, and WRH). CNVs were detected using PennCNV and QuantiSNP, with CNVRuler identifying 698 CNV regions (CNVRs), covering 109 Mb (4.83%) of the porcine genome. Comparison with previous CNV studies on swine revealed CNVR overlap rates ranging from 31.12% (French Yorkshire) to 81.27% (Xiang), and 9.06% newly identified CNVRs. DUC showed the most CNVRs (n = 384), followed by WRH (n = 225). Meanwhile, F1 and F2 exhibited far fewer CNVRs (five and seven, respectively). Functional enrichment analysis highlighted various genes overlapping with the CNVRs, including 1236 genes in DUC and 572 genes in WRH, linked to biological processes. The quantitative trait loci (QTLs), overlapping with CNVRs, exhibited particular overlapping with traits such as average daily gain (4.24% of QTLs in DUC, 4.51% of QTLs in WRH). In contrast, KNP, F1, and F2 populations exhibited a higher frequency of CNVRs containing QTLs overlapped with drip loss. These findings indicate that WRH may inherit growth traits from DUC. This study provides a better understanding of CNVs in the pigs, which can potentially be used in improving genetic merits of pig populations.
Collapse
Affiliation(s)
- Yong-Min Kim
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea; (Y.-M.K.); (Y.-S.K.); (J.-K.H.)
| | - Ha-Seung Seong
- Animal Breeding and Genetics Division, National Institute of Animal Science, Cheonan 31000, Republic of Korea;
| | - Seok-Joo Ha
- Department of Animal Science, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (S.-J.H.); (J.-K.K.); (H.B.); (S.K.); (W.-H.C.)
| | - Young-Sin Kim
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea; (Y.-M.K.); (Y.-S.K.); (J.-K.H.)
| | - Jae-Kwon Kim
- Department of Animal Science, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (S.-J.H.); (J.-K.K.); (H.B.); (S.K.); (W.-H.C.)
| | - Heejung Baek
- Department of Animal Science, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (S.-J.H.); (J.-K.K.); (H.B.); (S.K.); (W.-H.C.)
- Research Institute TNT Research Company, Jeonju 54810, Republic of Korea; (S.Y.); (D.S.)
| | - Seona Kwon
- Department of Animal Science, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (S.-J.H.); (J.-K.K.); (H.B.); (S.K.); (W.-H.C.)
| | - Sangwon Yoon
- Research Institute TNT Research Company, Jeonju 54810, Republic of Korea; (S.Y.); (D.S.)
| | - Joon-Hee Lee
- Department of Animal Bioscience, College of Agriculture and Life Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea;
| | - Dongwon Seo
- Research Institute TNT Research Company, Jeonju 54810, Republic of Korea; (S.Y.); (D.S.)
| | - Won-Hyong Chung
- Department of Animal Science, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (S.-J.H.); (J.-K.K.); (H.B.); (S.K.); (W.-H.C.)
| | - Joon-Ki Hong
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea; (Y.-M.K.); (Y.-S.K.); (J.-K.H.)
| | - Jung-Woo Choi
- Department of Animal Science, College of Animal Life Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea; (S.-J.H.); (J.-K.K.); (H.B.); (S.K.); (W.-H.C.)
| | - Eun-Seok Cho
- Swine Science Division, National Institute of Animal Science, Rural Development Administration, Cheonan 31000, Republic of Korea; (Y.-M.K.); (Y.-S.K.); (J.-K.H.)
| |
Collapse
|
16
|
Ament IH, DeBruyne N, Wang F, Lin L. Long-read RNA sequencing: A transformative technology for exploring transcriptome complexity in human diseases. Mol Ther 2025; 33:883-894. [PMID: 39563027 PMCID: PMC11897757 DOI: 10.1016/j.ymthe.2024.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/30/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
Long-read RNA sequencing (RNA-seq) is emerging as a powerful and versatile technology for studying human transcriptomes. By enabling the end-to-end sequencing of full-length transcripts, long-read RNA-seq opens up avenues for investigating various RNA species and features that cannot be reliably interrogated by standard short-read RNA-seq methods. In this review, we present an overview of long-read RNA-seq, delineating its strengths over short-read RNA-seq, as well as summarizing recent advances in experimental and computational approaches to boost the power of long-read-based transcriptomics. We describe a wide range of applications of long-read RNA-seq, and highlight its expanding role as a foundational technology for exploring transcriptome variations in human diseases.
Collapse
Affiliation(s)
| | - Nicole DeBruyne
- Graduate Group in Cell and Molecular Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Feng Wang
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| | - Lan Lin
- Center for Computational and Genomic Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
17
|
Guggenheim JA, Verhoeven VJM, Morgan IG. Myopia is predominantly genetic or predominantly environmental? Ophthalmic Physiol Opt 2025. [PMID: 40028922 DOI: 10.1111/opo.13464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 03/05/2025]
Affiliation(s)
| | - Virginie J M Verhoeven
- Department of Ophthalmology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Ian G Morgan
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
- Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
18
|
Mangnier L, Ruczinski I, Ricard J, Moreau C, Girard S, Maziade M, Bureau A. RetroFun-RVS: A Retrospective Family-Based Framework for Rare Variant Analysis Incorporating Functional Annotations. Genet Epidemiol 2025; 49:e70001. [PMID: 39876583 PMCID: PMC11775437 DOI: 10.1002/gepi.70001] [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: 04/03/2024] [Revised: 10/16/2024] [Accepted: 01/03/2025] [Indexed: 01/30/2025]
Abstract
A large proportion of genetic variations involved in complex diseases are rare and located within noncoding regions, making the interpretation of underlying biological mechanisms a daunting task. Although technical and methodological progress has been made to annotate the genome, current disease-rare-variant association tests incorporating such annotations suffer from two major limitations. First, they are generally restricted to case-control designs of unrelated individuals, which often require tens or hundreds of thousands of individuals to achieve sufficient power. Second, they were not evaluated with region-based annotations needed to interpret the causal regulatory mechanisms. In this work, we propose RetroFun-RVS, a new retrospective family-based score test, incorporating functional annotations. A critical feature of the proposed method is to aggregate genotypes to compare against rare variant-sharing expectations among affected family members. Through extensive simulations, we have demonstrated that RetroFun-RVS integrating networks based on 3D genome contacts as functional annotations reach greater power over the region-wide test, other strategies to include subregions and competing methods. Also, the proposed framework shows robustness to non-informative annotations, maintaining its power when causal variants are spread across regions. Asymptotic p-values are susceptible to Type I error inflation when the number of families with rare variants is small, and a bootstrap procedure is recommended in these instances. Application of RetroFun-RVS is illustrated on whole genome sequence in the Eastern Quebec Schizophrenia and Bipolar Disorder Kindred Study with networks constructed from 3D contacts and epigenetic data on neurons. In summary, the integration of functional annotations corresponding to regions or networks with transcriptional impacts in rare variant tests appears promising to highlight regulatory mechanisms involved in complex diseases.
Collapse
Affiliation(s)
- Loïc Mangnier
- Department of Social and Preventive MedicineLaval UniversityQuebec CityQuebecCanada
- CERVO Brain Research CenterQuebec CityQuebecCanada
- Big Data Research CenterLaval UniversityQuebec CityQuebecCanada
| | - Ingo Ruczinski
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | | | - Claudia Moreau
- Department of Fundamental SciencesUniversity of Quebec in ChicoutimiSaguenayQuebecCanada
| | - Simon Girard
- Department of Fundamental SciencesUniversity of Quebec in ChicoutimiSaguenayQuebecCanada
| | - Michel Maziade
- CERVO Brain Research CenterQuebec CityQuebecCanada
- Department of Psychiatry and NeurosciencesLaval UniversityQuebec CityQuebecCanada
| | - Alexandre Bureau
- Department of Social and Preventive MedicineLaval UniversityQuebec CityQuebecCanada
- CERVO Brain Research CenterQuebec CityQuebecCanada
- Big Data Research CenterLaval UniversityQuebec CityQuebecCanada
| |
Collapse
|
19
|
Gou Y, Jing Y, Wang Y, Li X, Yang J, Wang K, He H, Yang Y, Tang Y, Wang C, Xu J, Yang F, Li M, Tang Q. AnimalGWASAtlas: Annotation and prioritization of GWAS loci and quantitative trait loci for animal complex traits. J Biol Chem 2025; 301:108267. [PMID: 39909383 PMCID: PMC11904539 DOI: 10.1016/j.jbc.2025.108267] [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: 06/28/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/07/2025] Open
Abstract
Genome-wide association study (GWAS) and quantitative trait locus (QTL) mapping methods provide valuable insights and opportunities for identifying functional gene underlying phenotype formation. However, the majority of GWAS risk loci and QTLs located in noncoding regions poses significant challenges in pinpointing the protein-coding genes associated with specific traits. Moreover, growing evidence suggests not all GWAS risk loci and QTLs are functional, emphasizing the critical need for prioritizing causal sites-a task of paramount importance for biologists. The accumulation of publicly available multiomics data provides an unprecedented opportunity to annotate and prioritize GWAS risk loci and QTLs. Therefore, we developed a comprehensive multiomics database encompassing four major agricultural species-pig, sheep, cattle, and chicken. This database integrates publicly accessible datasets, including 140 GWAS studies (covering 471 traits), 2625 QTL datasets (spanning 1235 traits), 86 Hi-C datasets (from eight cells/tissue types), 95 epigenomic datasets (from four cells/tissue types), and 769 transcription factor motifs. The database aims to link GWAS-QTL loci located in the noncoding regions to the target genes they regulate and prioritize functional and causal regulatory elements. Ultimately, it provides a valuable resource and potential validation targets for elucidating the genes and molecular pathways underlying economically important traits in agricultural animals.
Collapse
Affiliation(s)
- Yuwei Gou
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yunhan Jing
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yifei Wang
- Department of Zoology, College of Life Science, Sichuan Agricultural University, Ya'an, Sichuan, China
| | - Xingyu Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jing Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Kai Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Hengdong He
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yuan Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yuanling Tang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Chen Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jun Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Fan Yang
- Mianyang Sanrun Cultural Technology Co, Ltd, Mianyang, Sichuan, China
| | - Mingzhou Li
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China.
| | - Qianzi Tang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China.
| |
Collapse
|
20
|
Valand A, Rajasekar P, Wain LV, Clifford RL. Interplay between genetics and epigenetics in lung fibrosis. Int J Biochem Cell Biol 2025; 180:106739. [PMID: 39848439 DOI: 10.1016/j.biocel.2025.106739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 12/15/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
Lung fibrosis, including idiopathic pulmonary fibrosis (IPF), is a complex and devastating disease characterised by the progressive scarring of lung tissue leading to compromised respiratory function. Aberrantly activated fibroblasts deposit extracellular matrix components into the surrounding lung tissue, impairing lung function and capacity for gas exchange. Both genetic and epigenetic factors have been found to play a role in the pathogenesis of lung fibrosis, with emerging evidence highlighting the interplay between these two regulatory mechanisms. This review provides an overview of the current understanding of the interplay between genetics and epigenetics in lung fibrosis. We discuss the genetic variants associated with susceptibility to lung fibrosis and explore how epigenetic modifications such as DNA methylation, histone modifications, and non-coding RNA expression contribute to disease. Insights from genome-wide association studies (GWAS) and epigenome-wide association studies (EWAS) are integrated to explore the molecular mechanisms underlying lung fibrosis pathogenesis. We also discuss the potential clinical implications of genetics and epigenetics in lung fibrosis, including the development of novel therapeutic targets. Overall, this review highlights the importance of considering both genetic and epigenetic factors in the understanding and management of lung fibrosis.
Collapse
Affiliation(s)
- Anita Valand
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, University of Nottingham, UK; Nottingham NIHR Biomedical Research Centre, Nottingham, UK; Biodiscovery Institute, University Park, University of Nottingham, UK
| | - Poojitha Rajasekar
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, University of Nottingham, UK; Nottingham NIHR Biomedical Research Centre, Nottingham, UK; Biodiscovery Institute, University Park, University of Nottingham, UK
| | - Louise V Wain
- Department of Population Health Sciences, University of Leicester, Leicester, UK; NIHR Leicester Biomedical Research Centre, University of Leicester, Leicester, UK
| | - Rachel L Clifford
- Centre for Respiratory Research, Translational Medical Sciences, School of Medicine, University of Nottingham, UK; Nottingham NIHR Biomedical Research Centre, Nottingham, UK; Biodiscovery Institute, University Park, University of Nottingham, UK.
| |
Collapse
|
21
|
Jahangiri Esfahani S, Ao X, Oveisi A, Diatchenko L. Rare variant association studies: Significance, methods, and applications in chronic pain studies. Osteoarthritis Cartilage 2025; 33:313-321. [PMID: 39725155 DOI: 10.1016/j.joca.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
Rare genetic variants, characterized by their low frequency in a population, have emerged as essential components in the study of complex disease genetics. The biology of rare variants underscores their significance, as they can exert profound effects on phenotypic variation and disease susceptibility. Recent advancements in sequencing technologies have yielded the availability of large-scale sequencing data such as the UK Biobank whole-exome sequencing (WES) cohort empowered researchers to conduct rare variant association studies (RVASs). This review paper discusses the significance of rare variants, available methodologies, and applications. We provide an overview of RVASs, emphasizing their relevance in unraveling the genetic architecture of complex diseases with special focus on chronic pain and Arthritis. Additionally, we discuss the strengths and limitations of various rare variant association testing methods, outlining a typical pipeline for conducting rare variant association. This pipeline encompasses crucial steps such as quality control of WES data, rare variant annotation, and association testing. It serves as a comprehensive guide for researchers in the field of chronic pain diseases interested in rare variant association studies in large-scale sequencing datasets like the UK Biobank WES cohort. Lastly, we discuss how the identified variants can be further investigated through detailed experimental studies in animal models to elucidate their functional impact and underlying mechanisms.
Collapse
Affiliation(s)
- Sahel Jahangiri Esfahani
- Faculty of Medicine and Health Sciences, Department of Human Genetics, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Xiang Ao
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Anahita Oveisi
- Department of Neuroscience, Faculty of Science, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Luda Diatchenko
- Faculty of Dental Medicine and Oral Health Sciences, Department of Anesthesia, Faculty of Medicine, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada.
| |
Collapse
|
22
|
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 2025; 26:156-170. [PMID: 39375560 DOI: 10.1038/s41576-024-00778-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 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
|
23
|
Wu Z, Zhao Y, Zhang B, Li Y. Causal association between serum 25-hydroxyvitamin D levels and epilepsy: A two-sample bidirectional mendelian randomization study. Epilepsy Behav 2025; 164:110253. [PMID: 39823739 DOI: 10.1016/j.yebeh.2024.110253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 12/25/2024] [Accepted: 12/28/2024] [Indexed: 01/20/2025]
Abstract
OBJECTIVE The study aimed to investigate the causal relationship between serum 25-hydroxyvitamin D (25(OH)D) levels and epilepsy using Mendelian randomization (MR), thereby addressing confounding and reverse causality issues in observational studies. METHODS We employed a two-sample bidirectional MR design utilizing summary-level data from the IEU OpenGWAS project. Serum 25(OH)D levels were analyzed using the publicly available dataset ebi-a-GCST90000618, which included 496,946 European samples and 68,960,93 SNPs. Data on epilepsy were obtained from ebi-a-GCST90018840, comprising 458,310 samples, including 4,382 epilepsy patients and 453,928 controls. To identify instrumental variables (IVs), we applied a significance threshold of P < 5e-8 for serum 25(OH)D levels as the exposure and P < 5e-6 for epilepsy as the exposure. IVs were required to demonstrate an r2 < 0.001 linkage disequilibrium and an F-statistic greater than 10. The MR analysis utilized five methods: inverse variance weighted (IVW), weighted median, MR-Egger, weighted mode, and simple mode, assessing causal relationships between serum 25(OH)D levels and epilepsy. Robustness checks included heterogeneity tests, leave-one-out sensitivity analyses, and assessments for horizontal pleiotropy. RESULTS Both directions of the MR analysis revealed no genetic correlation between serum 25(OH)D levels and epilepsy. CONCLUSION Our findings, supported by robust IV screening and consistent results across multiple MR methods, indicate a lack of causal relationship between serum 25(OH)D levels and epilepsy. These results suggest that while vitamin D plays a role in the nervous system, its relationship to epilepsy may not be direct, thus highlighting the need for further investigation in future studies.
Collapse
Affiliation(s)
- Zhanshen Wu
- Department of Pharmacy, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, China
| | - Yang Zhao
- Department of Pharmacy, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, China
| | - Bo Zhang
- Department of Pharmacy, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, China
| | - Yanyan Li
- Department of Pharmacy, the Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou 450015, China.
| |
Collapse
|
24
|
Kozlitina J, Sookoian S. Global Epidemiological Impact of PNPLA3 I148M on Liver Disease. Liver Int 2025; 45:e16123. [PMID: 39373119 DOI: 10.1111/liv.16123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/08/2024]
Abstract
The prevalence of metabolic dysfunction-associated steatotic liver disease (MASLD) has increased exponentially over the past three decades, in parallel with the global rise in obesity and type 2 diabetes. It is currently the most common cause of liver-related morbidity and mortality. Although obesity has been identified as a key factor in the increased prevalence of MASLD, individual differences in susceptibility are significantly influenced by genetic factors. PNPLA3 I148M (rs738409 C>G) is the variant with the greatest impact on the risk of developing progressive MASLD and likely other forms of steatotic liver disease. This variant is prevalent across the globe, with the risk allele (G) frequency exhibiting considerable variation. Here, we review the contribution of PNPLA3 I148M to global burden and regional differences in MASLD prevalence, focusing on recent evidence emerging from population-based sequencing studies and prevalence assessments. We calculated the population attributable fraction (PAF) as a means of quantifying the impact of the variant on MASLD. Furthermore, we employ quantitative trait locus (QTL) analysis to ascertain the associations between rs738409 and a range of phenotypic traits. This analysis suggests that these QTLs may underpin pleiotropic effects on extrahepatic traits. Finally, we outline potential avenues for further research and identify key areas for investigation in future studies.
Collapse
Affiliation(s)
- Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Silvia Sookoian
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Clinical and Molecular Hepatology, Translational Health Research Center (CENITRES), Maimónides University, Buenos Aires, Argentina
| |
Collapse
|
25
|
Fernandez TV. Genetics of Tourette Syndrome. Psychiatr Clin North Am 2025; 48:1-13. [PMID: 39880506 DOI: 10.1016/j.psc.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
This review explores the genetic basis of Tourette syndrome (TS), a complex neuropsychiatric disorder characterized by motor and vocal tics. Family, twin, and molecular genetic studies provide strong evidence for a genetic component in TS, with heritability estimates ranging from 50% to 80%. The genetic architecture of TS is complex, involving both common variants with small effects and rare variants with larger effects. Genetic studies have identified candidate genes and chromosomal regions associated with TS risk, implicating biological pathways related to neurodevelopment, neurotransmission, and synaptic function. The article also discusses the clinical implications of these findings and future research directions.
Collapse
Affiliation(s)
- Thomas V Fernandez
- Department of Psychiatry and Yale Child Study Center, Yale School of Medicine, 230 South Frontage Road, New Haven, CT 06520, USA.
| |
Collapse
|
26
|
May K, Hecker AS, Strube C, Yin T, König S. Genetic parameters and single-step genome-wide association analysis for trematode (Fasciola hepatica and Calicophoron/Paramphistomum spp.) infections in German dairy cows. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2025; 128:105712. [PMID: 39798592 DOI: 10.1016/j.meegid.2025.105712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/02/2025] [Accepted: 01/04/2025] [Indexed: 01/15/2025]
Abstract
Infections with the liver fluke (Fasciola hepatica) cause economic losses in cattle production worldwide. Also, infections with rumen flukes (Calicophoron/Paramphistomum spp.) are gaining importance in grazing cattle in Europe. However, increasing resistance of helminth parasites against anthelmintics and limitations in treatment emphasize the need for alternative breeding approaches. This study included 1602 dairy cows kept on 29 farms with 2423 observations for F. hepatica and Calicophoron/Paramphistomum spp. egg counts per gram faeces (EPG). The EPGs were binary defined (infected: EPG > 0; non-infected: EPG = 0) and logarithmically transformed. The pedigree included 7939 cows. Genotypes (777 k) were available for 214 cows. A single-step GBLUP (ssGBLUP) model was applied to estimate genetic parameters for infection traits. Genomic breeding values from ssGBLUP were used in a single-step genome-wide association study (ssGWAS) to identify genetic variants associated with helminth infections. The heritability for liver fluke infections was up to 0.09, and up to 0.34 for rumen fluke infections. The genetic correlations between liver and rumen fluke infections ranged from 0.49 to 0.53, indicating that breeding for improved resilience to both helminth taxa is possible simultaneously. The ssGWAS revealed four SNPs for liver fluke infections on BTA 5, 13, 26 and 29, and 17 SNPs for rumen fluke infections on BTA 3 and 23. The SNPs for liver fluke infections were annotated to 12 potential candidate genes, most of which involved in liver fibrosis and immunity. The LRRC8B gene was found to be involved in host-rumen fluke interactions.
Collapse
Affiliation(s)
- Katharina May
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, 30559 Hannover, Germany; Institute of Animal Breeding and Genetics, Justus-Liebig-University Gießen, 35390 Gießen, Germany.
| | - Anna Sophie Hecker
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Christina Strube
- Institute for Parasitology, Centre for Infection Medicine, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | - Tong Yin
- Institute of Animal Breeding and Genetics, Justus-Liebig-University Gießen, 35390 Gießen, Germany
| | - Sven König
- Institute of Animal Breeding and Genetics, Justus-Liebig-University Gießen, 35390 Gießen, Germany
| |
Collapse
|
27
|
Hill KR, Scelsi HF, Youngblood HA, Faralli JA, Itakura T, Fini ME, Peters DM, Lieberman RL. Structural basis for anomalous cellular trafficking behavior of glaucoma-associated A427T mutant myocilin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640437. [PMID: 40060664 PMCID: PMC11888440 DOI: 10.1101/2025.02.26.640437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Familial mutations in myocilin cause vision loss in glaucoma due to misfolding and a toxic gain of function in a senescent cell type in the anterior eye. Here we characterize the cellular behavior and structure of the myocilin (myocilin A427T) mutant, of uncertain pathogenicity. Our characterization of A427T demonstrates that even mutations that minimally perturb myocilin structure and stability can present challenges for protein quality control clearance pathways. Namely, when expressed in an inducible immortalized trabecular meshwork cell line, inhibition of the proteasome reroutes wild-type myocilin, but not myocilin A427T, from endoplasmic reticulum associated degradation to lysosomal degradation. Yet, the crystal structure of the A427T myocilin olfactomedin domain shows modest perturbations largely confined to the mutation site. The previously unappreciated range of mutant myocilin behavior correlating with variable stability and structure provides a rationale for why it is challenging to predict causal pathogenicity of a given myocilin mutation, even in the presence of clinical data for members of an affected family. Comprehending the continuum of mutant myocilin behavior in the laboratory supports emerging efforts to use genetics to assess glaucoma risk in the clinic. In addition, the study supports a therapeutic strategy aimed at enhancing autophagic clearance of mutant myocilin.
Collapse
Affiliation(s)
- Kamisha R Hill
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA
| | - Hailee F Scelsi
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA
| | - Hannah A Youngblood
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA
| | - Jennifer A Faralli
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI
| | - Tatsuo Itakura
- USC Institute for Genetic Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA
| | - M Elizabeth Fini
- New England Eye Center, Tufts Medical Center; Department of Ophthalmology, School of Medicine and Tufts Graduate School of Biomedical Sciences, Tufts University, Boston, MA
| | - Donna M Peters
- Department of Pathology & Laboratory Medicine, University of Wisconsin, Madison, WI
| | - Raquel L Lieberman
- School of Chemistry & Biochemistry, Georgia Institute of Technology, Atlanta, GA
| |
Collapse
|
28
|
Ruffo P, Traynor BJ, Conforti FL. Advancements in genetic research and RNA therapy strategies for amyotrophic lateral sclerosis (ALS): current progress and future prospects. J Neurol 2025; 272:233. [PMID: 40009238 PMCID: PMC11865122 DOI: 10.1007/s00415-025-12975-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
This review explores the intricate landscape of neurodegenerative disease research, focusing on Amyotrophic Lateral Sclerosis (ALS) and the intersection of genetics and RNA biology to investigate the causative pathogenetic basis of this fatal disease. ALS is a severe neurodegenerative disease characterized by the progressive loss of motor neurons, leading to muscle weakness and paralysis. Despite significant research advances, the exact cause of ALS remains largely unknown. Thanks to the application of next-generation sequencing (NGS) approaches, it was possible to highlight the fundamental role of rare variants with large effect sizes and involvement of portions of non-coding RNA, providing valuable information on risk prediction, diagnosis, and treatment of age-related diseases, such as ALS. Genetic research has provided valuable insights into the pathophysiology of ALS, leading to the development of targeted therapies such as antisense oligonucleotides (ASOs). Regulatory agencies in several countries are evaluating the commercialization of Qalsody (Tofersen) for SOD1-associated ALS, highlighting the potential of gene-targeted therapies. Furthermore, the emerging significance of microRNAs (miRNAs) and long RNAs are of great interest. MiRNAs have emerged as promising biomarkers for diagnosing ALS and monitoring disease progression. Understanding the role of lncRNAs in the pathogenesis of ALS opens new avenues for therapeutic intervention. However, challenges remain in delivering RNA-based therapeutics to the central nervous system. Advances in genetic screening and personalized medicine hold promise for improving the management of ALS. Ongoing clinical trials use genomic approaches for patient stratification and drug targeting. Further research into the role of non-coding RNAs in the pathogenesis of ALS and their potential as therapeutic targets is crucial to the development of effective treatments for this devastating disease.
Collapse
Affiliation(s)
- Paola Ruffo
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA.
- Medical Genetics Laboratory, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Bryan J Traynor
- Neuromuscular Diseases Research Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Francesca Luisa Conforti
- Medical Genetics Laboratory, Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| |
Collapse
|
29
|
He Z, Chu B, Yang J, Gu J, Chen Z, Liu L, Morrison T, Belloy ME, Qi X, Hejazi N, Mathur M, Le Guen Y, Tang H, Hastie T, Ionita-laza I, Candès E, Sabatti C. Beyond guilty by association at scale: searching for causal variants on the basis of genome-wide summary statistics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.02.28.582621. [PMID: 38464202 PMCID: PMC10925326 DOI: 10.1101/2024.02.28.582621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Understanding the causal genetic architecture of complex phenotypes will fuel future research into disease mechanisms and potential therapies. Here, we illustrate the power of a novel framework: it detects, starting from summary statistics, and across the entire genome, sets of variants that carry non-redundant information on the phenotypes and are therefore more likely to be causal in a biological sense. The approach, implemented in open-source software, is also computationally efficient, requiring less than 15 minutes on a single CPU to perform genome-wide analysis. Through extensive genome-wide simulation studies, we show that the method can substantially outperform existing methods in false discovery rate control, statistical power and various fine-mapping criteria. In applications to a meta-analysis of ten large-scale genetic studies of Alzheimer's disease (AD), we identified 82 loci associated with AD, including 37 additional loci missed by conventional GWAS pipeline. Massively parallel reporter assays and CRISPR-Cas9 experiments have confirmed the functionality of the putative causal variants our method points to. Finally, we retrospectively analyzed summary statistics from 67 large-scale GWAS for a variety of phenotypes. Results reveal the method's capacity to robustly discover additional loci for polygenic traits and pinpoint potential causal variants underpinning each locus beyond conventional GWAS pipeline, contributing to a deeper understanding of complex genetic architectures in post-GWAS analyses.
Collapse
Affiliation(s)
- Zihuai He
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - Benjamin Chu
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
| | - James Yang
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Jiaqi Gu
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Zhaomeng Chen
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Linxi Liu
- Department of Statistics, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Tim Morrison
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Michael E. Belloy
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Xinran Qi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Nima Hejazi
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Maya Mathur
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Yann Le Guen
- Quantitative Sciences Unit, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Hua Tang
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Trevor Hastie
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| | - Iuliana Ionita-laza
- Department of Biostatistics, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Emmanuel Candès
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
- Department of Mathematics, Stanford University, Stanford, CA 94305, USA
| | - Chiara Sabatti
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Statistics, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
30
|
Zhang C, Yu H, Miao Y, Wei B. Causal relationship between osteoporosis, bone mineral density, and osteonecrosis: a bidirectional two-sample Mendelian randomization study. J Transl Med 2025; 23:226. [PMID: 40001090 PMCID: PMC11863788 DOI: 10.1186/s12967-024-06030-9] [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: 08/13/2024] [Accepted: 12/25/2024] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Osteonecrosis (ON) is a debilitating orthopedic condition characterized by bone cell death due to impaired blood supply, leading to structural changes and disability. Osteoporosis (OP), a systemic skeletal disease, results in reduced bone density and quality, making bones fragile and prone to fractures. Although distinct, OP and ON share several risk factors such as corticosteroid use and smoking. This study aims to investigate the causal relationships between OP, bone mineral density (BMD), and ON using a bidirectional two-sample Mendelian randomization (MR) approach. METHODS This study utilized genome-wide association study (GWAS) data for OP from the FinnGen database, and BMD data for the lumbar spine and femoral neck from the Genetic Factors for Osteoporosis (GEFOS) consortium. ON data were also obtained from the FinnGen database. All participants were of European descent. Genetic instruments were selected based on genome-wide significance, linkage disequilibrium, and strength (F-statistic). Bidirectional MR analysis was performed using inverse-variance weighted (IVW), MR-Egger regression, and weighted median methods to assess causality. Sensitivity analyses, including Cochran's Q test and MR-PRESSO, were conducted to evaluate heterogeneity and pleiotropy. RESULTS MR analysis demonstrated a positive causal effect of OP on ON using the IVW method, with an odds ratio (OR) of 1.223 (95% CI: 1.026-1.459, P = 0.025). The weighted median method also confirmed this result with an OR (95% CI) 1.290 (1.021-1.630), P = 0.033. No significant causal effects were found between BMD (lumbar spine and femoral neck) and ON. Furthermore, ON did not exhibit a causal effect on OP or BMD. Sensitivity analyses confirmed the robustness of the results, showing no evidence of heterogeneity or pleiotropy. CONCLUSION This study provides evidence of a unidirectional causal relationship between OP and ON, suggesting that individuals with a genetic predisposition to OP have an increased risk of developing ON. These findings highlight the importance of early OP detection and management to potentially reduce ON incidence. The lack of a significant causal relationship between BMD and ON indicates that factors other than bone density, such as vascular health, may play a crucial role in ON development. Future research should explore these mechanisms further to inform clinical interventions.
Collapse
Affiliation(s)
- Chao Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hao Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yulin Miao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | | |
Collapse
|
31
|
Rout M, Ramu D, Mariana M, Koshy T, Venkatesan V, Lopez-Alvarenga JC, Arya R, Ravichandran U, Sharma SK, Lodha S, Ponnala AR, Sharma KK, Shaik MV, Resendez RG, Venugopal P, R P, S N, Ezeilo JA, Almeida M, Paralta J, Mummidi S, Natesan C, Mehra NK, Singh JR, Wander GS, Ralhan S, Blackett PR, Blangero J, Medicherla KM, Thanikachalam S, Panchatcharam TS, K DK, Gupta R, Paul SFD, Ghosh AK, Aston CE, Duggirala R, Sanghera DK. Excess of rare noncoding variants in several type 2 diabetes candidate genes among Asian Indian families. COMMUNICATIONS MEDICINE 2025; 5:47. [PMID: 39987249 PMCID: PMC11846969 DOI: 10.1038/s43856-025-00750-9] [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: 11/28/2023] [Accepted: 01/23/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) etiology is highly complex due to its multiple roots of origin. Polygenic risk scores (PRS) based on genome-wide association studies (GWAS) can partially explain T2D risk. Asian Indian people have up to six times higher risk of developing T2D than European people, and underlying causes of this disparity are unknown. METHODS We have performed targeted sequencing of ten T2D GWAS/candidate regions using endogamous Punjabi Sikh families and replication studies using unrelated Sikh people and families from three other Indian endogamous ethnic groups (EEGs). RESULTS We detect rare and ultra-rare variants (RVs) in KCNJ11-ABCC8 and HNF4A (MODY genes) cosegregated with late-onset T2D. We also identify RV enrichment in two new genes, SLC38A11 and ANPEP, associated with T2D. Gene-burden analysis reveals the highest RV burden contributed by HNF4A (p = 0.0003), followed by KCNJ11/ABCC8 (p = 0.0061) and SLC38A11 (p = 0.03). Some RVs detected in Sikh people are also found in Agarwals from Jaipur, both from Northern India, but were monomorphic in other two EEGs from South Indian people. Despite carrying a high burden of T2D and RVs, most families have a significantly lower burden of PRS. Functional studies show that an intronic regulatory variant (RV) in ABCC8 affects the binding of Pax4 and NF-kB transcription factors, influencing downstream gene regulation. CONCLUSIONS The high burden of T2D in these families may stem from the enrichment of noncoding RVs in a small number of major known genes (including MODY genes) with oligogenic inheritance alongside RVs from genes associated with polygenic susceptibility. These findings highlight the need to conduct deeper evaluations of families from non-European ancestries to identify potential novel therapeutics and implement preventative strategies.
Collapse
Affiliation(s)
- Madhusmita Rout
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Deepika Ramu
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Mendez Mariana
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Teena Koshy
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Vettriselvi Venkatesan
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Juan C Lopez-Alvarenga
- Department of Population Health & Biostatistics, University of Texas Rio Grande Valley (UTRGV), Harlingen, TX, USA
| | - Rector Arya
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Umarani Ravichandran
- Department of Medicine, Rajah Muthiah Medical College Hospital, Annamalai University, Chidambaram, India
| | | | - Sailesh Lodha
- Departments of Preventive Cardiology, Internal Medicine and Endocrinology, Eternal Heart Care Centre and Research Institute, Mount Sinai New York Affiliate, Jaipur, India
| | - Amaresh Reddy Ponnala
- Department of Endocrinology, Krishna Institute of Medical Sciences (KIMS) Hospital, Nellore, India
| | - Krishna Kumar Sharma
- Department of Pharmacology, Lal Bahadur Shastri College of Pharmacy, Rajasthan University of Health Sciences, Jaipur, India
| | - Mahaboob Vali Shaik
- Department of Endocrinology, Narayana Medical College and Hospital, Nellore, India
| | - Roy G Resendez
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Priyanka Venugopal
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Parthasarathy R
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Noelta S
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Juliet A Ezeilo
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Marcio Almeida
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley (UTRGV), Brownsville, TX, USA
| | - Juan Paralta
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley (UTRGV), Brownsville, TX, USA
| | - Srinivas Mummidi
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Chidambaram Natesan
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Narinder K Mehra
- All India Institute of Medical Sciences and Research, New Delhi, India
| | | | | | - Sarju Ralhan
- Hero Dayanand Medical College and Heart Institute, Ludhiana, India
| | - Piers R Blackett
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley (UTRGV), Brownsville, TX, USA
| | | | - Sadagopan Thanikachalam
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Thyagarajan Sadras Panchatcharam
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Dileep Kumar K
- Department of Endocrinology, Narayana Medical College and Hospital, Nellore, India
| | - Rajeev Gupta
- Departments of Preventive Cardiology, Internal Medicine and Endocrinology, Eternal Heart Care Centre and Research Institute, Mount Sinai New York Affiliate, Jaipur, India
| | - Solomon Franklin D Paul
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research (Deemed to be University), Chennai, India
| | - Asish K Ghosh
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Christopher E Aston
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ravindranath Duggirala
- Department of Health and Behavioral Sciences, Texas A&M University-San Antonio, San Antonio, TX, US
| | - Dharambir K Sanghera
- Department of Pediatrics, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Pharmaceutical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Physiology, College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
32
|
Hoffmann M, Hennighausen L. Spotlight on amino acid changing mutations in the JAK-STAT pathway: from disease-specific mutation to general mutation databases. Sci Rep 2025; 15:6202. [PMID: 39979591 PMCID: PMC11842829 DOI: 10.1038/s41598-025-90788-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
The JAK-STAT pathway is central to cytokine signaling and controls normal physiology and disease. Aberrant activation via mutations that change amino acids in proteins of the pathway can result in diseases. While disease-centric databases like COSMIC catalog mutations in cancer, their prevalence in healthy populations remains underexplored. We systematically studied such mutations in the JAK-STAT genes by comparing COSMIC and the population-focused All of Us database. Our analysis revealed frequent mutations in all JAK and STAT domains, particularly among white females. We further identified three categories: Mutations uniquely found in All of Us that were associated with cancer in the literature but could not be found in COSMIC, underscoring COSMIC's limitations. Mutations unique to COSMIC underline their potential as drivers of cancer due to their absence in the general population. Mutations present in both databases, e.g., JAK2Val617Phe/V617F - widely recognized as a cancer driver in hematopoietic cells, but without disease associations in All of Us, raising the possibility that combinatorial SNPs might be responsible for disease development. These findings illustrate the complementarity of both databases for understanding mutation impacts and underscore the need for multi-mutation analyses to uncover genetic factors underlying complex diseases and advance personalized medicine.
Collapse
Affiliation(s)
- Markus Hoffmann
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA.
| | - Lothar Hennighausen
- Laboratory of Genetics and Physiology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, 20892, USA
| |
Collapse
|
33
|
Iyer KR, Clarke SL, Guarischi-Sousa R, Gjoni K, Heath AS, Young EP, Stitziel NO, Laurie C, Broome JG, Khan AT, Lewis JP, Xu H, Montasser ME, Ashley KE, Hasbani NR, Boerwinkle E, Morrison AC, Chami N, Do R, Rocheleau G, Lloyd-Jones DM, Lemaitre RN, Bis JC, Floyd JS, Kinney GL, Bowden DW, Palmer ND, Benjamin EJ, Nayor M, Yanek LR, Kral BG, Becker LC, Kardia SLR, Smith JA, Bielak LF, Norwood AF, Min YI, Carson AP, Post WS, Rich SS, Herrington D, Guo X, Taylor KD, Manson JE, Franceschini N, Pollard KS, Mitchell BD, Loos RJF, Fornage M, Hou L, Psaty BM, Young KA, Regan EA, Freedman BI, Vasan RS, Levy D, Mathias RA, Peyser PA, Raffield LM, Kooperberg C, Reiner AP, Rotter JI, Jun G, de Vries PS, Assimes TL. Unveiling the Genetic Landscape of Coronary Artery Disease Through Common and Rare Structural Variants. J Am Heart Assoc 2025; 14:e036499. [PMID: 39950338 DOI: 10.1161/jaha.124.036499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/21/2024] [Indexed: 02/17/2025]
Abstract
BACKGROUND Genome-wide association studies have identified several hundred susceptibility single nucleotide variants for coronary artery disease (CAD). Despite single nucleotide variant-based genome-wide association studies improving our understanding of the genetics of CAD, the contribution of structural variants (SVs) to the risk of CAD remains largely unclear. METHOD AND RESULTS We leveraged SVs detected from high-coverage whole genome sequencing data in a diverse group of participants from the National Heart Lung and Blood Institute's Trans-Omics for Precision Medicine program. Single variant tests were performed on 58 706 SVs in a study sample of 11 556 CAD cases and 42 907 controls. Additionally, aggregate tests using sliding windows were performed to examine rare SVs. One genome-wide significant association was identified for a common biallelic intergenic duplication on chromosome 6q21 (P=1.54E-09, odds ratio=1.34). The sliding window-based aggregate tests found 1 region on chromosome 17q25.3, overlapping USP36, to be significantly associated with coronary artery disease (P=1.03E-10). USP36 is highly expressed in arterial and adipose tissues while broadly affecting several cardiometabolic traits. CONCLUSIONS Our results suggest that SVs, both common and rare, may influence the risk of coronary artery disease.
Collapse
Affiliation(s)
- Kruthika R Iyer
- Data Science and Biotechnology, Gladstone Institutes San Francisco CA USA
- Department of Medicine, Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA USA
| | - Shoa L Clarke
- Department of Medicine, Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA USA
- Department of Medicine, Stanford Prevention Research Center Stanford University School of Medicine Stanford CA USA
| | - Rodrigo Guarischi-Sousa
- Department of Medicine, Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA USA
| | - Ketrin Gjoni
- Data Science and Biotechnology, Gladstone Institutes San Francisco CA USA
- Department of Epidemiology and Biostatistics University of California San Francisco CA USA
| | - Adam S Heath
- Department of Epidemiology, Human Genetics Center, School of Public Health The University of Texas Health Science Center at Houston Houston TX USA
| | - Erica P Young
- Department of Medicine, Division of Cardiology Washington University School of Medicine Saint Louis MO USA
- McDonnell Genome Institute, Washington University School of Medicine Saint Louis MO USA
| | - Nathan O Stitziel
- Department of Medicine, Division of Cardiology Washington University School of Medicine Saint Louis MO USA
- McDonnell Genome Institute, Washington University School of Medicine Saint Louis MO USA
- Department of Genetics Washington University School of Medicine Saint Louis MO USA
| | - Cecelia Laurie
- Department of Biostatistics University of Washington Seattle WA USA
| | - Jai G Broome
- Department of Biostatistics University of Washington Seattle WA USA
- Department of Medicine, Division of Internal Medicine University of Washington Seattle WA USA
| | - Alyna T Khan
- Department of Biostatistics University of Washington Seattle WA USA
| | - Joshua P Lewis
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Huichun Xu
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - May E Montasser
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA
| | - Kellan E Ashley
- Department of Medicine University of Mississippi Medical Center Jackson MS USA
| | - Natalie R Hasbani
- Department of Epidemiology, Human Genetics Center, School of Public Health The University of Texas Health Science Center at Houston Houston TX USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics Center, School of Public Health The University of Texas Health Science Center at Houston Houston TX USA
- Human Genome Sequencing Center Baylor College of Medicine Houston TX USA
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics Center, School of Public Health The University of Texas Health Science Center at Houston Houston TX USA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized Medicine Icahn School of Medicine at Mount Sinai New York NY USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine Icahn School of Medicine at Mount Sinai New York NY USA
| | - Ghislain Rocheleau
- The Charles Bronfman Institute for Personalized 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
| | | | - Rozenn N Lemaitre
- Department of Medicine, Cardiovascular Health Research Unit University of Washington Seattle WA USA
| | - Joshua C Bis
- Department of Medicine, Cardiovascular Health Research Unit University of Washington Seattle WA USA
| | - James S Floyd
- Department of Medicine, Cardiovascular Health Research Unit University of Washington Seattle WA USA
- Department of Epidemiology University of Washington Seattle WA USA
| | - Gregory L Kinney
- Department of Epidemiology Colorado School of Public Health Aurora CO USA
| | - Donald W Bowden
- Department of Biochemistry Wake Forest University School of Medicine Winston-Salem NC USA
| | - Nicholette D Palmer
- Department of Biochemistry Wake Forest University School of Medicine Winston-Salem NC USA
| | - Emelia J Benjamin
- Department of Medicine, Cardiovascular Medicine, Boston Medical Center Boston University Chobanian & Avedisian School of Medicine Boston MA USA
- Department of Epidemiology Boston University School of Public Health Boston MA USA
| | - Matthew Nayor
- Department of Medicine, Cardiovascular Medicine Boston University Chobanian & Avedisian School of Medicine Boston MA USA
- Department of Medicine, Preventive Medicine & Epidemiology Boston University Chobanian & Avedisian School of Medicine Boston MA USA
| | - Lisa R Yanek
- Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Brian G Kral
- Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Lewis C Becker
- Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Sharon L R Kardia
- Department of Epidemiology University of Michigan School of Public Health Ann Arbor MI USA
| | - Jennifer A Smith
- Department of Epidemiology University of Michigan School of Public Health Ann Arbor MI USA
- Institute for Social Research Survey Research Center, University of Michigan Ann Arbor MI USA
| | - Lawrence F Bielak
- Department of Epidemiology University of Michigan School of Public Health Ann Arbor MI USA
| | - Arnita F Norwood
- Department of Medicine University of Mississippi Medical Center Jackson MS USA
| | - Yuan-I Min
- Department of Medicine University of Mississippi Medical Center Jackson MS USA
| | - April P Carson
- Department of Medicine University of Mississippi Medical Center Jackson MS USA
| | - Wendy S Post
- Department of Medicine, Division of Cardiology Johns Hopkins University Baltimore MD USA
| | - Stephen S Rich
- Department of Genome Sciences University of Virginia School of Medicine Charlottesville VA USA
| | - David Herrington
- Department of Medicine Wake Forest University School of Medicine Winston-Salem NC USA
| | - Xiuqing Guo
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center Torrance CA USA
| | - Kent D Taylor
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center Torrance CA USA
| | - JoAnn E Manson
- Department of Medicine Brigham and Women's Hospital, Harvard Medical School Boston MA USA
| | - Nora Franceschini
- Department of Epidemiology University of North Carolina at Chapel Hill Chapel Hill NC USA
| | - Katherine S Pollard
- Data Science and Biotechnology, Gladstone Institutes San Francisco CA USA
- Department of Epidemiology and Biostatistics University of California San Francisco CA USA
- Chan Zuckerberg Biohub San Francisco CA USA
| | - Braxton D Mitchell
- Department of Medicine University of Maryland School of Medicine Baltimore MD USA
- Geriatric Research and Education Clinical Center Baltimore Veterans Administration Medical Center Baltimore MD USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine Icahn School of Medicine at Mount Sinai New York NY USA
- Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Basic Metabolic Research University of Copenhagen Copenhagen Denmark
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine University of Texas Health Science Center at Houston Houston TX USA
| | - Lifang Hou
- Department of Preventive Medicine Northwestern University Chicago IL USA
| | - Bruce M Psaty
- Department of Medicine, Cardiovascular Health Research Unit University of Washington Seattle WA USA
- Department of Epidemiology University of Washington Seattle WA USA
- Department of Health Systems and Population Health University of Washington Seattle WA USA
| | - Kendra A Young
- Department of Epidemiology Colorado School of Public Health Aurora CO USA
| | | | - Barry I Freedman
- Department of Internal Medicine, Section on Nephrology Wake Forest University School of Medicine Winston-Salem NC USA
| | | | - Daniel Levy
- Division of Intramural Research, Population Sciences Branch National Heart, Lung, and Blood Institute, National Institutes of Health Bethesda MD USA
| | - Rasika A Mathias
- Department of Medicine Johns Hopkins University School of Medicine Baltimore MD USA
| | - Patricia A Peyser
- Department of Epidemiology University of Michigan School of Public Health Ann Arbor MI USA
| | - Laura M Raffield
- Department of Genetics University of North Carolina at Chapel Hill Chapel Hill NC USA
| | | | - Alex P Reiner
- Division of Public Health Fred Hutchinson Cancer Center Seattle WA USA
| | - Jerome I Rotter
- Department of Pediatrics, The Institute for Translational Genomics and Population Sciences The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center Torrance CA USA
| | - Goo Jun
- Department of Epidemiology, Human Genetics Center, School of Public Health The University of Texas Health Science Center at Houston Houston TX USA
| | - Paul S de Vries
- Department of Epidemiology, Human Genetics Center, School of Public Health The University of Texas Health Science Center at Houston Houston TX USA
| | - Themistocles L Assimes
- Department of Medicine, Division of Cardiovascular Medicine Stanford University School of Medicine Stanford CA USA
- VA Palo Alto Healthcare System Palo Alto CA USA
| |
Collapse
|
34
|
Li D, Tian Y, Vona B, Yu X, Lin J, Ma L, Lou S, Li X, Zhu G, Wang Y, Du M, Wang L, Pan Y. A TAF11 variant contributes to non-syndromic cleft lip only through modulating neural crest cell migration. Hum Mol Genet 2025; 34:392-401. [PMID: 39727181 DOI: 10.1093/hmg/ddae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 09/30/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024] Open
Abstract
The NC_000006.12: g.34887814C>G variant in TAF11 was identified as a potential functional variant in a Chinese pedigree including two non-syndromic cleft lip only (NSCLO) cases. Applying Chromatin Immunoprecipitation (ChIP), Electrophoretic mobility shift and super-shift assays, we found that the mutant G allele recruited more STAT1 and STAT3, and increased the expression of TAF11. RNA sequencing, GO and KEGG pathway enrichment, ChIP and dual-luciferase reporter assays revealed that TAF11 downregulated CDH1 and CTNND1 in the cell adhesion pathway by binding to their promoter regions and inhibiting transcriptional activities. Alcian blue staining, time-lapse photography, whole-mount in situ hybridization, phospho-Histone H3 immunofluorescence and TUNEL assays indicated that TAF11 and taf11 overexpression (TAF11OE and taf11OE, respectively) contributed to disturbed migration of cranial neural crest cells and abnormal craniofacial development, as well as increased death and deformity rates in zebrafish. In conclusion, a functionally relevant TAF11 variant, affecting cell migration via modulating CDH1 and CTNND1, was associated with etiology of NSCLO.
Collapse
Affiliation(s)
- Dandan Li
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, No. 1 Shanghai Road, Gulou District, Nanjing 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
| | - Yu Tian
- Department of Stomatology, Zhenjiang First People's Hospital, People's Hospital Affiliated to Jiangsu University, No. 8 Electric Road, Runzhou District, Zhenjiang 212000, China
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen 37075, Germany
- Institute for Auditory Neuroscience and Inner Ear Lab, University Medical Center Göttingen, Robert-Koch-Str. 40, Göttingen 37075, Germany
| | - Xin Yu
- Department of Orthodontics, Affiliated Nantong Stomatological Hospital of Nantong University, No. 36 Yuelong South Road, Chongchuan District, Nantong 226006, China
| | - Junyan Lin
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, No. 1 Shanghai Road, Gulou District, Nanjing 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
| | - Lan Ma
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
| | - Shu Lou
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, No. 1 Shanghai Road, Gulou District, Nanjing 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
| | - Xiaofeng Li
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, No. 1 Shanghai Road, Gulou District, Nanjing 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
| | - Guirong Zhu
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
| | - Yuting Wang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
| | - Mulong Du
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, No. 101 Longmian Avenue, Jiangning District, Nanjing 211166, China
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, No. 101 Longmian Avenue, Jiangning District, Nanjing 211166, China
| | - Lin Wang
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, No. 1 Shanghai Road, Gulou District, Nanjing 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
| | - Yongchu Pan
- Department of Orthodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, No. 1 Shanghai Road, Gulou District, Nanjing 210029, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, No. 136 Hanzhong Road, Gulou District, Nanjing 210029, China
| |
Collapse
|
35
|
Zhang Y, Hu T, Wang X, Sun N, Cai Q, Kim HY, Fan Y, Liu D, Guan X. Profiles of gut microbiota and metabolites for high risk of transgenerational depression-like behavior by paternal methamphetamine exposure. FASEB J 2025; 39:e70386. [PMID: 39927989 DOI: 10.1096/fj.202402839r] [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: 11/13/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Parental substance abuse increases the risk of neurological and psychiatric disorders in offsprings. However, its underlying mechanism remains elusive. Our previous study demonstrated that long-term exposure to methamphetamine (Meth), a psychostimulant drug with high addiction potential, remarkably alters the gut microbiome and metabolites in male mice, which contribute to Meth-induced anxiety-like behaviors. The current study aimed to investigate whether gut microbiota and metabolism serve as potential peripheral targets for transgenerational mental problems by paternal Meth exposure. We found that paternal Meth exposure induced depression-like behaviors both in the first (F1) and the second (F2) generations of male mice. Further, the depletion of gut bacteria through antibiotic treatments normalized the depression-like behaviors to normal levels in both F1 and F2 male mice. Then, alterations in gut bacterial composition were observed in both F1 and F2 male mice. Specifically, Eubacterium_ruminantium_group, Enterorhabdus, Alloprevotella, and Parabacteroides were the commonly affected bacterial taxa in F1 and F2 male mice. In addition, the results of alterations in gut metabolism showed that LPC 14:1-SN1 emerged as the consistently altered metabolite in the colons of F1 and F2 male mice. Taken together, our findings provide the first evidence that paternal Meth exposure enhances depression-like behaviors in F1 and F2 male mice, potentially mediated by the gut microbiome and metabolism.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tao Hu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xinyu Wang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Nongyuan Sun
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qinglong Cai
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hee Young Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dekang Liu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
36
|
Azam S, Sahu A, Pandey NK, Neupane M, Van Tassell CP, Rosen BD, Gandham RK, Rath SN, Majumdar SS. Advancing the Indian cattle pangenome: characterizing non-reference sequences in Bos indicus. J Anim Sci Biotechnol 2025; 16:21. [PMID: 39915889 PMCID: PMC11804092 DOI: 10.1186/s40104-024-01133-1] [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: 08/15/2024] [Accepted: 11/26/2024] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND India harbors the world's largest cattle population, encompassing over 50 distinct Bos indicus breeds. This rich genetic diversity underscores the inadequacy of a single reference genome to fully capture the genomic landscape of Indian cattle. To comprehensively characterize the genomic variation within Bos indicus and, specifically, dairy breeds, we aim to identify non-reference sequences and construct a comprehensive pangenome. RESULTS Five representative genomes of prominent dairy breeds, including Gir, Kankrej, Tharparkar, Sahiwal, and Red Sindhi, were sequenced using 10X Genomics 'linked-read' technology. Assemblies generated from these linked-reads ranged from 2.70 Gb to 2.77 Gb, comparable to the Bos indicus Brahman reference genome. A pangenome of Bos indicus cattle was constructed by comparing the newly assembled genomes with the reference using alignment and graph-based methods, revealing 8 Mb and 17.7 Mb of novel sequence respectively. A confident set of 6,844 Non-reference Unique Insertions (NUIs) spanning 7.57 Mb was identified through both methods, representing the pangenome of Indian Bos indicus breeds. Comparative analysis with previously published pangenomes unveiled 2.8 Mb (37%) commonality with the Chinese indicine pangenome and only 1% commonality with the Bos taurus pangenome. Among these, 2,312 NUIs encompassing ~ 2 Mb, were commonly found in 98 samples of the 5 breeds and designated as Bos indicus Common Insertions (BICIs) in the population. Furthermore, 926 BICIs were identified within 682 protein-coding genes, 54 long non-coding RNAs (lncRNA), and 18 pseudogenes. These protein-coding genes were enriched for functions such as chemical synaptic transmission, cell junction organization, cell-cell adhesion, and cell morphogenesis. The protein-coding genes were found in various prominent quantitative trait locus (QTL) regions, suggesting potential roles of BICIs in traits related to milk production, reproduction, exterior, health, meat, and carcass. Notably, 63.21% of the bases within the BICIs call set contained interspersed repeats, predominantly Long Interspersed Nuclear Elements (LINEs). Additionally, 70.28% of BICIs are shared with other domesticated and wild species, highlighting their evolutionary significance. CONCLUSIONS This is the first report unveiling a robust set of NUIs defining the pangenome of Bos indicus breeds of India. The analyses contribute valuable insights into the genomic landscape of desi cattle breeds.
Collapse
Affiliation(s)
- Sarwar Azam
- National Institute of Animal Biotechnology, Hyderabad, India
- Indian Institute of Technology Hyderabad, Sangareddy, India
| | - Abhisek Sahu
- National Institute of Animal Biotechnology, Hyderabad, India
| | | | - Mahesh Neupane
- Animal Genomics and Improvement Laboratory, USDA-ARS, Beltsville, MD, 20705, USA
| | - Curtis P Van Tassell
- Animal Genomics and Improvement Laboratory, USDA-ARS, Beltsville, MD, 20705, USA
| | - Benjamin D Rosen
- Animal Genomics and Improvement Laboratory, USDA-ARS, Beltsville, MD, 20705, USA.
| | - Ravi Kumar Gandham
- National Institute of Animal Biotechnology, Hyderabad, India.
- Animal Biotechnology, ICAR-NBAGR, Karnal, Haryana, India.
| | | | | |
Collapse
|
37
|
Zhang X, Wu W, Zhou G, Huang X, Xu M, Zhao Q, Yan H. Relationship between alcohol use and traumatic brain injury: evidence from Mendelian randomization. Brain Inj 2025:1-8. [PMID: 39894956 DOI: 10.1080/02699052.2025.2460740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND Observational studies suggest that alcohol consumption increases the risk of traumatic brain injury (TBI); however, the causality of this association remains unclear. OBJECTIVES This study aimed to identify which drinking pattern is the primary factor influencing TBI. METHOD Two-sample Mendelian randomization (MR) was used to assess whether drinking patterns (alcohol consumption, abuse, and intake frequency) are causally associated with TBI risk. RESULTS MR analysis revealed causal effects of alcohol intake frequency [odds ratio (OR) 0.806, 95% confidence interval (CI): 0.665-0.978, p = 0.028, beta: -0.215, se: 0.098], alcohol drinks per week (OR 1.772, 95% CI: 1.140-2.753, p = 0.011, beta: 0.572, se: 0.225), and alcohol abuse (OR 1.095, 95% CI: 1.006-1.192, p = 0.035, beta: 0.091, se: 0.043) on TBI. Additionally, no causal effect of alcohol consumption (OR 0.730, 95% CI: 0.264-2.025, p = 0.546, beta: -0.314, se: 0.520) or average monthly alcohol intake (OR 1.138, 95% CI: 0.805-1.609, p = 0.463, beta: 0.130, se: 0.177) on TBI was observed. Similarly, the effects of TBI on alcohol intake were statistically non-significant. CONCLUSION Drinking patterns, including alcohol intake frequency and abuse, influence TBI, whereas TBI rarely influences drinking patterns.
Collapse
Affiliation(s)
- Xiaohang Zhang
- School of Integrated Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenze Wu
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guisheng Zhou
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resource Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xi Huang
- School of Integrated Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Xu
- School of Integrated Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiulong Zhao
- Department of Pharmacy, Shenzhen Hospital of Integrated Traditional Chinese and Western Medicine, Shenzhen, China
| | - Hui Yan
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
38
|
Belot A, Tusseau M, Cognard J, Georgin‐Lavialle S, Boursier G, Hedrich CM. How (Ultra-)Rare Gene Variants Improve Our Understanding of More Common Autoimmune and Inflammatory Diseases. ACR Open Rheumatol 2025; 7:e70003. [PMID: 39964335 PMCID: PMC11834591 DOI: 10.1002/acr2.70003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/21/2025] Open
Abstract
The aim of this study was to explore the impact of rare and ultra-rare genetic variants on the understanding and treatment of autoimmune and autoinflammatory diseases with a focus on systemic lupus erythematosus (SLE) and Behçet syndrome. This review summarizes current research on the monogenic causes of SLE and Behçet syndrome, highlighting the various pathways that can be responsible for these unique phenotypes. In monogenic SLE, the identification of complement and DNASE1L3 deficiencies has elucidated mechanisms of apoptotic body accumulation and extracellular nucleic acid sensing. Type I interferonopathies underline the specific role of DNA/RNA sensing and the interferon overexpression in the development of systemic autoimmunity. Other significant genetic defects include Toll-like receptor hypersignaling and JAK/STATopathies, which contribute to the breakdown of immune tolerance. To date, genetic defects directly affecting B and T cell biology only account for a minority of identified causes of monogenic lupus, highlighting the importance of a tight regulation of mechanistic target of rapamycin and RAS (Rat sarcoma GTPase)/MAPK (mitogen-activated protein kinase) signaling in lupus. In Behçet syndrome, rare variants in TNFAIP3, RELA, and NFKB1 genes have been identified, underscoring the importance of NF-κB overactivation. Additional monogenic diseases such as ELF4, WDR1 mutations and trisomy 8 further illustrate the genetic complexity of this condition. Observations from genetic studies in SLE and Behçet syndrome highlight the complexity of systemic inflammatory diseases in which distinct molecular defects caused by single-gene mutations can promote lupus or Behçet syndromes, often unrecognizable from their genetically complex "classical" forms. Insights gained from studying rare genetic variants enhance our understanding of immune function in health and disease, paving the way for targeted therapies and personalized medicine.
Collapse
Affiliation(s)
- Alexandre Belot
- Centre International de Recherche en Infectiologie, University of Lyon, Inserm U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, École normale supérieure de Lyon, National Referee Centre for Rheumatic and Autoimmune and Systemic Diseases in Children, and Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Lyon, France, and French National Reference Center of Autoinflammatory Diseases and AmyloidosisLyonFrance
| | - Maud Tusseau
- Centre International de Recherche en Infectiologie, University of Lyon, Inserm U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, École normale supérieure de Lyon, National Referee Centre for Rheumatic and AutoImmune and Systemic Diseases in Children, and Hôpital Femme Mère Enfant and Groupement Hospitalier Est, Hospices Civils de Lyon, Lyon, France, and French National Reference Center of Autoinflammatory Diseases and AmyloidosisParisFrance
| | - Jade Cognard
- American Memorial Hospital, Centre Hospitalier Universitaire Reims, Reims Champagne‐Ardenne UniversityReimsFrance
| | - Sophie Georgin‐Lavialle
- French National Reference Center of Autoinflammatory Diseases and Amyloidosis, Paris, France, and Sorbonne Université, Hôpital Tenon, DMU 3ID, AP‐HPParisFrance
| | - Guilaine Boursier
- French National Reference Center of Autoinflammatory Diseases and Amyloidosis, Paris, France, and Centre Hospitalier Universitaire Montpellier, University of MontpellierMontpellierFrance
| | - Christian M. Hedrich
- Institute of Life Course and Medical Sciences, University of Liverpool and Alder Hey Children's NHS Foundation TrustLiverpoolUnited Kingdom
| |
Collapse
|
39
|
Moore L, Malde S, Dasgupta P, Sahai A, Raison N. Inheritance patterns of lower urinary tract symptoms in adults: a systematic review. BJU Int 2025; 135:192-203. [PMID: 39187949 PMCID: PMC11745988 DOI: 10.1111/bju.16517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
OBJECTIVE To compile and evaluate the heritability and inheritance patterns of lower urinary tract symptoms (LUTS) in adult cohorts. METHODS Searches of five databases (PubMed, Embase, APA PsycInfo, Global Health, and OVID Medline) commenced on 6 July 2024, resulting in 736 articles retrieved after deduplication. Studies evaluating heritability patterns, gene frequencies, and familial aggregation of symptoms were included for review. Screening and predefined eligibility criteria produced 34 studies for final review. A descriptive analysis of synthesised data was performed, adhering to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. The Cochrane Risk of Bias in Non-Randomised Studies of Interventions (ROBINS-I) tool and the Johanna Briggs Institute checklist were used to evaluate these studies. RESULTS Ten of the 34 studies (29%) described general LUTS, 14 (41%) described symptoms due to benign prostatic enlargement (BPE), nine (26%) described urinary incontinence (UI; urge UI [UUI], stress UI [SUI] and mixed UI [MUI]), four (12%) described nocturia alone, two (6%) described overactive bladder (OAB), and four (13%) described other specific symptoms (frequency, postvoid residual urine volume). BPE symptoms, UI (MUI and UUI), nocturia alone, and frequency alone were associated with genetic predisposition, whilst OAB and SUI had more modest inheritance. CONCLUSION The pathogenetic and pharmacological mechanisms fundamental to LUTS manifestation are highly heterogeneous. Further work is required to evaluate the inheritance patterns of LUTS more extensively.
Collapse
Affiliation(s)
- Lorcan Moore
- Guy's King's and St Thomas' School of Medical EducationKing's College LondonLondonUK
| | - Sachin Malde
- Department of UrologyGuy's and St Thomas' HospitalLondonUK
| | - Prokar Dasgupta
- Guy's King's and St Thomas' School of Medical EducationKing's College LondonLondonUK
- Department of UrologyGuy's and St Thomas' HospitalLondonUK
| | - Arun Sahai
- Department of UrologyGuy's and St Thomas' HospitalLondonUK
| | - Nicholas Raison
- Guy's King's and St Thomas' School of Medical EducationKing's College LondonLondonUK
- Department of UrologyKing's College HospitalLondonUK
| |
Collapse
|
40
|
Koido M. Polygenic modelling and machine learning approaches in pharmacogenomics: Importance in downstream analysis of genome-wide association study data. Br J Clin Pharmacol 2025; 91:264-269. [PMID: 37743713 PMCID: PMC11773102 DOI: 10.1111/bcp.15913] [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: 07/31/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 09/26/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified genetic variations associated with adverse drug effects in pharmacogenomics (PGx) research. However, interpreting the biological implications of these associations remains a challenge. This review highlights 2 promising post-GWAS methods for PGx. First, we discuss the polygenic architecture of the PGx traits, especially for drug-induced liver injury. Experimental modelling using multiple donors' human primary hepatocytes and human liver organoids demonstrated the polygenic architecture of drug-induced liver injury susceptibility and found biological vulnerability in genetically high-risk tissue donors. Second, we discuss the challenges of interpreting the roles of variants in noncoding regions. Beyond methods involving expression quantitative trait locus analysis and massively parallel reporter assays, we suggest the use of in silico mutagenesis through machine learning methods to understand the roles of variants in transcriptional regulation. This review underscores the importance of these post-GWAS methods in providing critical insights into PGx, potentially facilitating drug development and personalized treatment.
Collapse
Affiliation(s)
- Masaru Koido
- Laboratory of Complex Trait Genomics, Department of Computational Biology and Medical Sciences, Graduate School of Frontier SciencesThe University of TokyoTokyoJapan
| |
Collapse
|
41
|
Alemu R, Sharew NT, Arsano YY, Ahmed M, Tekola-Ayele F, Mersha TB, Amare AT. Multi-omics approaches for understanding gene-environment interactions in noncommunicable diseases: techniques, translation, and equity issues. Hum Genomics 2025; 19:8. [PMID: 39891174 PMCID: PMC11786457 DOI: 10.1186/s40246-025-00718-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
Non-communicable diseases (NCDs) such as cardiovascular diseases, chronic respiratory diseases, cancers, diabetes, and mental health disorders pose a significant global health challenge, accounting for the majority of fatalities and disability-adjusted life years worldwide. These diseases arise from the complex interactions between genetic, behavioral, and environmental factors, necessitating a thorough understanding of these dynamics to identify effective diagnostic strategies and interventions. Although recent advances in multi-omics technologies have greatly enhanced our ability to explore these interactions, several challenges remain. These challenges include the inherent complexity and heterogeneity of multi-omic datasets, limitations in analytical approaches, and severe underrepresentation of non-European genetic ancestries in most omics datasets, which restricts the generalizability of findings and exacerbates health disparities. This scoping review evaluates the global landscape of multi-omics data related to NCDs from 2000 to 2024, focusing on recent advancements in multi-omics data integration, translational applications, and equity considerations. We highlight the need for standardized protocols, harmonized data-sharing policies, and advanced approaches such as artificial intelligence/machine learning to integrate multi-omics data and study gene-environment interactions. We also explore challenges and opportunities in translating insights from gene-environment (GxE) research into precision medicine strategies. We underscore the potential of global multi-omics research in advancing our understanding of NCDs and enhancing patient outcomes across diverse and underserved populations, emphasizing the need for equity and fairness-centered research and strategic investments to build local capacities in underrepresented populations and regions.
Collapse
Affiliation(s)
- Robel Alemu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Anderson School of Management, University of California Los Angeles, Los Angeles, CA, USA.
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| | - Nigussie T Sharew
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Yodit Y Arsano
- Alpert Medical School, Lifespan Health Systems, Brown University, WarrenProvidence, Rhode Island, USA
| | - Muktar Ahmed
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Population Health Research, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Tesfaye B Mersha
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| | - Azmeraw T Amare
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia.
| |
Collapse
|
42
|
Dawoody Nejad L, Pioro EP. Modeling ALS with Patient-Derived iPSCs: Recent Advances and Future Potentials. Brain Sci 2025; 15:134. [PMID: 40002468 PMCID: PMC11852857 DOI: 10.3390/brainsci15020134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a terminal complex neurodegenerative disease, with 10-15% of cases being familial and the majority being sporadic with no known cause. There are no animal models for the 85-90% of sporadic ALS cases. More creative, sophisticated models of ALS disease are required to unravel the mysteries of this complicated disease. While ALS patients urgently require new medications and treatments, suitable preclinical in vitro models for drug screening are lacking. Therefore, human-derived induced pluripotent stem cell (hiPSC) technology offers the opportunity to model diverse and unreachable cell types in a culture dish. In this review, we focus on recent hiPSC-derived ALS neuronal and non-neuronal models to examine the research progress of current ALS 2D monocultures, co-cultures, and more complex 3D-model organoids. Despite the challenges inherent to hiPSC-based models, their application to preclinical drug studies is enormous.
Collapse
Affiliation(s)
| | - Erik P. Pioro
- Djavad Mowafaghian Centre for Brain Health, Division of Neurology, Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
| |
Collapse
|
43
|
López-Catalina A, Ragab M, Reverter A, González-Recio O. A Recursive Model Approach to Include Epigenetic Effects in Genetic Evaluations Using Simulated DNA Methylation Effects. J Anim Breed Genet 2025. [PMID: 39868874 DOI: 10.1111/jbg.12925] [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: 09/06/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/28/2025]
Abstract
The advancement of epigenetics has highlighted DNA methylation as an intermediate-omic influencing gene regulation and phenotypic expression. With emerging technologies enabling the large-scale and affordable capture of methylation data, there is growing interest in integrating this information into genetic evaluation models for animal breeding. This study used methylome information from six dairy cows to simulate the methylation profile of 13,183 genotyped animals. The liability to methylation was treated as an additive trait, while a trait moderated by methylation effects was also simulated. A multiomic model (GOBLUP) was adapted to incorporate methylation data in genomic and genetic evaluations, using the traditional BLUP method as a benchmark. The GOBLUP accurately recovered heritability estimates for the liability to methylation in all low, medium and high heritability scenarios and was consistent at estimating the heritability for the epigenetics-moderated trait of interest at a low-medium heritability of 0.14. The genetic variance recovered by the BLUP model was influenced by the h2 of the liability to methylation, and a part of the methylation variance for the phenotypic trait was captured as additive. The h2 of the phenotypic trait partially relies on the h2 value for the methylation windows in the traditional model. A newly proposed estimated epigenetic value (EEV) combines the traditional additive genetic information from genotyping arrays with epigenetic information. The correlation between the traditional estimated breeding value (EBV) and EEV was high (0.92-0.99 depending on the scenario), but the correlation of the EEV with the true breeding value was higher than the correlation between the traditional EBV and the TBV (0.85 vs. 0.75, 0.71 vs. 0.66 and 0.61 vs. 0.62 depending on the scenario). This study demonstrates that the GOBLUP multiomic recursive model can effectively separates additive and epigenetic variances, enabling improved breeding decisions by accounting for genetic liability to DNA methylation. This enables more informed breeding decisions, optimising selection for desired traits. Emerging sequencing techniques offer new opportunities for cost-effective simultaneous acquisition of genetic and epigenetic data, further enhancing breeding accuracy.
Collapse
Affiliation(s)
- Adrián López-Catalina
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), CSIC, Madrid, Spain
- Departamento de Producción Agraria, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid, Ciudad Universitaria s/n, Madrid, Spain
- CSIRO Agriculture & Food, Queensland Bioscience Precinct, Brisbane, Queensland, Australia
| | - Mohamed Ragab
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), CSIC, Madrid, Spain
| | - Antonio Reverter
- CSIRO Agriculture & Food, Queensland Bioscience Precinct, Brisbane, Queensland, Australia
| | - Oscar González-Recio
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), CSIC, Madrid, Spain
| |
Collapse
|
44
|
Ikeda I, Igarashi R, Fujihara K, Takeda Y, Ferreira ED, Mon KL, Kodama S, Mori Y, Kadowaki T, Honda R, Arase Y, Sone H. Cross-sectional and Longitudinal Associations Between Family History of Type 2 Diabetes Mellitus, Hypertension, and Dyslipidemia and Their Prevalence and Incidence: Toranomon Hospital Health Management Center Study (TOPICS24). Mayo Clin Proc 2025:S0025-6196(24)00615-3. [PMID: 39895435 DOI: 10.1016/j.mayocp.2024.10.020] [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: 04/17/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 02/04/2025]
Abstract
OBJECTIVE To examine the association between a positive family history (parents, siblings, and grandparents) of type 2 diabetes mellitus (T2DM), hypertension, and dyslipidemia and their prevalence and incidence in the same population. PATIENTS AND METHODS Data on 41,361 participants who underwent health examinations between January 1, 1997, and December 31, 2007, were analyzed, and the results of logistic and Cox regression analyses in the same cohort were examined. RESULTS Cross-sectional analyses showed that the prevalence of all three diseases increased with a positive family history, especially T2DM, with an odds ratio (OR) of 12.00 (95% CI, 7.82 to 18.41) when the number of affected relatives was greater than or equal to 3 with an OR of 20.43 (95% CI, 11.0 to 37.8) for a positive family history across three generations compared with no family history. However, redefining family history from "parents, siblings, and grandparents" to "parents and siblings" or "parents only" did not significantly change ORs for each disease. Among those with a positive family history and body mass index greater than or equal to 30.0 kg/m2 hypertension was 19 times more prevalent compared with no family history and body mass index of 18.5 to 24.9 kg/m2. In the longitudinal study, family history strongly influenced incident T2DM (hazard ratio[HR], 2.40; 95% CI, 1.93 to 2.98), hypertension (HR, 1.43; 95% CI, 1.26 to 1.62), and dyslipidemia (HR, 1.41; 95% CI, 1.08 to 1.83), respectively. CONCLUSION Obtaining a family history of these diseases was useful in identifying high-risk groups. Also, for T2DM, the influence of a positive family history was strongest with a marked increase in risk with overlap of affected family members, suggesting that a family history is useful for early detection and prevention.
Collapse
Affiliation(s)
- Izumi Ikeda
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Risa Igarashi
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Kazuya Fujihara
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Yasunaga Takeda
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Efrem d'Ávila Ferreira
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Khin Lay Mon
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Satoru Kodama
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Yasumichi Mori
- Department of Endocrinology and Metabolism, Toranomon Hospital, Tokyo, Japan
| | | | - Ritsuko Honda
- Health Management Center, Toranomon Hospital, Tokyo, Japan
| | - Yasuji Arase
- Health Management Center, Toranomon Hospital, Tokyo, Japan
| | - Hirohito Sone
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan.
| |
Collapse
|
45
|
Lee CL, Chuang CK, Chiu HC, Chang YH, Tu YR, Lo YT, Lin HY, Lin SP. Understanding Genetic Screening: Harnessing Health Information to Prevent Disease Risks. Int J Med Sci 2025; 22:903-919. [PMID: 39991772 PMCID: PMC11843151 DOI: 10.7150/ijms.101219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 12/17/2024] [Indexed: 02/25/2025] Open
Abstract
Genetic screening analyzes an individual's genetic information to assess disease risk and provide personalized health recommendations. This article introduces the public to genetic screening, explaining its definition, principles, history, and common types, including prenatal, newborn, adult disease risk, cancer, and pharmacogenetic screening. It elaborates on the benefits of genetic screening, such as early risk detection, personalized prevention, family risk assessment, and reproductive decision-making. The article also notes limitations, including result interpretation uncertainty, psychological and ethical issues, and privacy and discrimination risks. It provides advice on selecting suitable screening, consulting professionals, choosing reliable institutions, and understanding screening purposes and limitations. Finally, it discusses applying screening results through lifestyle adjustments, regular check-ups, and preventive treatments. By comprehensively introducing genetic screening, the article aims to raise public awareness and encourage utilizing this technology to prevent disease and maintain health.
Collapse
Affiliation(s)
- Chung-Lin Lee
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Chih-Kuang Chuang
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
| | - Huei-Ching Chiu
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
| | - Ya-Hui Chang
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yuan-Rong Tu
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Yun-Ting Lo
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
| | - Hsiang-Yu Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Mackay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Shuan-Pei Lin
- Department of Pediatrics, MacKay Memorial Hospital, Taipei, Taiwan
- International Rare Disease Center, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
- Division of Genetics and Metabolism, Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Department of Infant and Child Care, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| |
Collapse
|
46
|
Jung S, Caballero M, Olfson E, Newcorn JH, Fernandez TV, Mahjani B. Rare Variant Analyses in Ancestrally Diverse Cohorts Reveal Novel ADHD Risk Genes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.14.25320294. [PMID: 39867378 PMCID: PMC11759603 DOI: 10.1101/2025.01.14.25320294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly heritable neurodevelopmental disorder, but its genetic architecture remains incompletely characterized. Rare coding variants, which can profoundly impact gene function, represent an underexplored dimension of ADHD risk. In this study, we analyzed large-scale DNA sequencing datasets from ancestrally diverse cohorts and observed significant enrichment of rare protein-truncating and deleterious missense variants in highly evolutionarily constrained genes. This analysis identified 15 high-confidence ADHD risk genes, including the previously implicated KDM5B. Integrating these findings with genome-wide association study (GWAS) data revealed nine enriched pathways, with strong involvement in synapse organization, neuronal development, and chromatin regulation. Protein-protein interaction analyses identified chromatin regulators as central network hubs, and single-cell transcriptomic profiling confirmed their expression in neurons and glial cells, with distinct patterns in oligodendrocyte subtypes. These findings advance our understanding of the genetic architecture of ADHD, uncover core molecular mechanisms, and provide promising directions for future therapeutic development.
Collapse
Affiliation(s)
- Seulgi Jung
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Madison Caballero
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Olfson
- Child Study Center, Yale University, New Haven, CT, USA
- Wu Tsai Institute, Yale University, New Haven, CT, USA
| | - Jeffrey H. Newcorn
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas V. Fernandez
- Child Study Center, Yale University, New Haven, CT, USA
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Behrang Mahjani
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, 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
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
47
|
Martins Rodrigues F, Jasielec J, Perpich M, Kim A, Moma L, Li Y, Storrs E, Wendl MC, Jayasinghe RG, Fiala M, Stefka A, Derman B, Jakubowiak AJ, DiPersio JF, Vij R, Godley LA, Ding L. Germline predisposition in multiple myeloma. iScience 2025; 28:111620. [PMID: 39845416 PMCID: PMC11750583 DOI: 10.1016/j.isci.2024.111620] [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: 09/08/2024] [Revised: 10/04/2024] [Accepted: 11/14/2024] [Indexed: 01/24/2025] Open
Abstract
We present a study of rare germline predisposition variants in 954 unrelated individuals with multiple myeloma (MM) and 82 MM families. Using a candidate gene approach, we identified such variants across all age groups in 9.1% of sporadic and 18% of familial cases. Implicated genes included genes suggested in other MM risk studies as potential risk genes (DIS3, EP300, KDM1A, and USP45); genes involved in predisposition to other cancers (ATM, BRCA1/2, CHEK2, PMS2, POT1, PRF1, and TP53); and BRIP1, EP300, and FANCM in individuals of African ancestry. Variants were characterized using loss of heterozygosity (LOH), biallelic events, and gene expression analyses, revealing 31 variants in 3.25% of sporadic cases for which pathogenicity was supported by multiple lines of evidence. Our results suggest that the disruption of DNA damage repair pathways may play a role in MM susceptibility. These results will inform improved surveillance in high-risk groups and potential therapeutic strategies.
Collapse
Affiliation(s)
- Fernanda Martins Rodrigues
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jagoda Jasielec
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Melody Perpich
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Aelin Kim
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Luke Moma
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Yize Li
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erik Storrs
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Michael C. Wendl
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Reyka G. Jayasinghe
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark Fiala
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew Stefka
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Benjamin Derman
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Andrzej J. Jakubowiak
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - John F. DiPersio
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ravi Vij
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lucy A. Godley
- Division of Hematology/Oncology, Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Li Ding
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Division of Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
48
|
Castanho I, Yeganeh PN, Boix CA, Morgan SL, Mathys H, Prokopenko D, White B, Soto LM, Pegoraro G, Shah S, Ploumakis A, Kalavros N, Bennett DA, Lange C, Kim DY, Bertram L, Tsai LH, Kellis M, Tanzi RE, Hide W. Molecular hallmarks of excitatory and inhibitory neuronal resilience and resistance to Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.13.632801. [PMID: 39868232 PMCID: PMC11761133 DOI: 10.1101/2025.01.13.632801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Background A significant proportion of individuals maintain healthy cognitive function despite having extensive Alzheimer's disease (AD) pathology, known as cognitive resilience. Understanding the molecular mechanisms that protect these individuals can identify therapeutic targets for AD dementia. This study aims to define molecular and cellular signatures of cognitive resilience, protection and resistance, by integrating genetics, bulk RNA, and single-nucleus RNA sequencing data across multiple brain regions from AD, resilient, and control individuals. Methods We analyzed data from the Religious Order Study and the Rush Memory and Aging Project (ROSMAP), including bulk (n=631) and multi-regional single nucleus (n=48) RNA sequencing. Subjects were categorized into AD, resilient, and control based on β-amyloid and tau pathology, and cognitive status. We identified and prioritized protected cell populations using whole genome sequencing-derived genetic variants, transcriptomic profiling, and cellular composition distribution. Results Transcriptomic results, supported by GWAS-derived polygenic risk scores, place cognitive resilience as an intermediate state in the AD continuum. Tissue-level analysis revealed 43 genes enriched in nucleic acid metabolism and signaling that were differentially expressed between AD and resilience. Only GFAP (upregulated) and KLF4 (downregulated) showed differential expression in resilience compared to controls. Cellular resilience involved reorganization of protein folding and degradation pathways, with downregulation of Hsp90 and selective upregulation of Hsp40, Hsp70, and Hsp110 families in excitatory neurons. Excitatory neuronal subpopulations in the entorhinal cortex (ATP8B1+ and MEF2Chigh) exhibited unique resilience signaling through neurotrophin (modulated by LINGO1) and angiopoietin (ANGPT2/TEK) pathways. We identified MEF2C, ATP8B1, and RELN as key markers of resilient excitatory neuronal populations, characterized by selective vulnerability in AD. Protective rare variant enrichment highlighted vulnerable populations, including somatostatin (SST) inhibitory interneurons, validated through immunofluorescence showing co-expression of rare variant associated RBFOX1 and KIF26B in SST+ neurons in the dorsolateral prefrontal cortex. The maintenance of excitatory-inhibitory balance emerges as a key characteristic of resilience. Conclusions We identified molecular and cellular hallmarks of cognitive resilience, an intermediate state in the AD continuum. Resilience mechanisms include preservation of neuronal function, maintenance of excitatory/inhibitory balance, and activation of protective signaling pathways. Specific excitatory neuronal populations appear to play a central role in mediating cognitive resilience, while a subset of vulnerable SST interneurons likely provide compensation against AD-associated dysregulation. This study offers a framework to leverage natural protective mechanisms to mitigate neurodegeneration and preserve cognition in AD.
Collapse
Affiliation(s)
- Isabel Castanho
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Pourya Naderi Yeganeh
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Carles A. Boix
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sarah L. Morgan
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Centre for Neuroscience, Surgery and Trauma, Blizard Institute, Queen Mary University of London, London E1 2AT, UK
| | - Hansruedi Mathys
- University of Pittsburgh Brain Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
| | - Dmitry Prokopenko
- Harvard Medical School, Boston, MA, USA
- Genetics and Aging Research Unit, The Henry and Allison McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Bartholomew White
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Larisa M. Soto
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Giulia Pegoraro
- Harvard Medical School, Boston, MA, USA
- Medical School, University of Exeter, Exeter EX2 5DW, UK
| | | | - Athanasios Ploumakis
- Harvard Medical School, Boston, MA, USA
- Spatial Technologies Unit, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nikolas Kalavros
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David A. Bennett
- Rush Alzheimer’s Disease Center, Rush University Medical Center, 1750 W Harrison Street, Suite 1000, Chicago, IL, 60612, USA
| | - Christoph Lange
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, 02115, Boston, MA, USA
| | - Doo Yeon Kim
- Harvard Medical School, Boston, MA, USA
- Genetics and Aging Research Unit, The Henry and Allison McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Lars Bertram
- Lübeck Interdisciplinary Platform for Genome Analytics, Institutes of Neurogenetics and Cardiogenetics, University of Lübeck, Lübeck, Germany
- Department of Psychology, University of Oslo, Oslo, Norway
| | - Li-Huei Tsai
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Picower Institute for Learning and Memory, MIT, Cambridge, MA 02139, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA 02139, USA
| | - Manolis Kellis
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rudolph E. Tanzi
- Harvard Medical School, Boston, MA, USA
- Genetics and Aging Research Unit, The Henry and Allison McCance Center for Brain Health, Department of Neurology, Massachusetts General Hospital, Boston, MA, United States
| | - Winston Hide
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| |
Collapse
|
49
|
Wu XR, Yang L, Wu BS, Liu WS, Deng YT, Kang JJ, Dong Q, Sahakian BJ, Feng JF, Cheng W, Yu JT. Exome sequencing identifies genes for socioeconomic status in 350,770 individuals. Proc Natl Acad Sci U S A 2025; 122:e2414018122. [PMID: 39772748 PMCID: PMC11745334 DOI: 10.1073/pnas.2414018122] [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: 07/12/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Socioeconomic status (SES) is a critical factor in determining health outcomes and is influenced by genetic and environmental factors. However, our understanding of the genetic structure of SES remains incomplete. Here, we conducted a large-scale exome study of SES markers (household income, occupational status, educational attainment, and social deprivation) in 350,770 individuals. For rare coding variants, we identified 56 significant associations by gene-based collapsing tests, unveiling 7 additional SES-associated genes (NRN1, CCDC36, RHOB, EP400, NCAM1, TPTEP2-CSNK1E, and LINC02881). Exome-wide single common variant analysis revealed nine lead single-nucleotide polymorphisms (SNPs) associated with household income and 34 lead SNPs associated with EduYears, replicating previous GWAS findings. The gene-environment correlations had a substantial impact on the genetic associations with SES, as indicated by the significantly increased P values in several associations after controlling for geographic regions. Furthermore, we observed the pleiotropic effects of SES-associated genetic factors on a wide range of health outcomes, such as cognitive function, psychosocial status, and diabetes. This study highlights the contribution of coding variants to SES and their associations with health phenotypes.
Collapse
Affiliation(s)
- Xin-Rui Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Liu Yang
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Bang-Sheng Wu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Wei-Shi Liu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Yue-Ting Deng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Ju-Jiao Kang
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai200433, China
| | - Qiang Dong
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| | - Barbara J. Sahakian
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai200433, China
- Department of Psychiatry and Behavioural and Clinical Neuroscience Institute, University of Cambridge, CambridgeCB2 0SZ, United Kingdom
| | - Jian-Feng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai200433, China
- Department of Computer Science, University of Warwick, CoventryCV4 7AL, United Kingdom
| | - Wei Cheng
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai200433, China
| | - Jin-Tai Yu
- Department of Neurology and National Center for Neurological Disorders, Huashan Hospital, Fudan University, Shanghai200040, China
- State Key Laboratory of Medical Neurobiology and Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai200040, China
| |
Collapse
|
50
|
Zhang D, Gao B, Feng Q, Manichaikul A, Peloso GM, Tracy RP, Durda P, Taylor KD, Liu Y, Johnson WC, Gabriel S, Gupta N, Smith JD, Aguet F, Ardlie KG, Blackwell TW, Gerszten RE, Rich SS, Rotter JI, Scott LJ, Zhou X, Lee S. Proteome-wide association studies for blood lipids and comparison with transcriptome-wide association studies. HGG ADVANCES 2025; 6:100383. [PMID: 39543875 PMCID: PMC11650301 DOI: 10.1016/j.xhgg.2024.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/17/2024] Open
Abstract
Blood lipid traits are treatable and heritable risk factors for heart disease, a leading cause of mortality worldwide. Although genome-wide association studies (GWASs) have discovered hundreds of variants associated with lipids in humans, most of the causal mechanisms of lipids remain unknown. To better understand the biological processes underlying lipid metabolism, we investigated the associations of plasma protein levels with total cholesterol (TC), triglycerides (TG), high-density lipoprotein (HDL) cholesterol, and low-density lipoprotein (LDL) cholesterol in blood. We trained protein prediction models based on samples in the Multi-Ethnic Study of Atherosclerosis (MESA) and applied them to conduct proteome-wide association studies (PWASs) for lipids using the Global Lipids Genetics Consortium (GLGC) data. Of the 749 proteins tested, 42 were significantly associated with at least one lipid trait. Furthermore, we performed transcriptome-wide association studies (TWASs) for lipids using 9,714 gene expression prediction models trained on samples from peripheral blood mononuclear cells (PBMCs) in MESA and 49 tissues in the Genotype-Tissue Expression (GTEx) project. We found that although PWASs and TWASs can show different directions of associations in an individual gene, 40 out of 49 tissues showed a positive correlation between PWAS and TWAS signed p values across all the genes, which suggests high-level consistency between proteome-lipid associations and transcriptome-lipid associations.
Collapse
Affiliation(s)
- Daiwei Zhang
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA; Departments of Biostatistics and Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Boran Gao
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Qidi Feng
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Gina M Peloso
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Russell P Tracy
- Departments of Pathology and Laboratory Medicine, and Biochemistry, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Peter Durda
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Yongmei Liu
- Department of Medicine, Divisions of Cardiology and Neurology, Duke University Medical Center, Durham, NC, USA
| | - W Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Stacey Gabriel
- Genomics Platform, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Namrata Gupta
- Genomics Platform, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Joshua D Smith
- Department of Genome Sciences, Human Genetics, and Translational Genomics, University of Washington, Seattle, WA, USA
| | - Francois Aguet
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Kristin G Ardlie
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Thomas W Blackwell
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Laura J Scott
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA.
| | - Xiang Zhou
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Seunggeun Lee
- Graduate School of Data Science, Seoul National University, Seoul, Republic of Korea; Department of Biostatistics and Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|