1
|
Shan Y, Hu H, Yang A, Zhao W, Chu Y. An integrative approach to identifying NPC1 as a susceptibility gene for gestational diabetes mellitus. J Matern Fetal Neonatal Med 2025; 38:2445665. [PMID: 39746811 DOI: 10.1080/14767058.2024.2445665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/24/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVE The objective of this study was to identify a novel gene and its potential mechanisms associated with susceptibility to gestational diabetes mellitus (GDM) through an integrative approach. METHODS We analyzed data from genome-wide association studies (GWAS) of GDM in the FinnGen R11 dataset (16,802 GDM cases and 237,816 controls) and Genotype Tissue Expression v8 expression quantitative trait locus data. We used summary-data-based Mendelian randomization to determine associations between transcript levels and phenotypes, transcriptome-wide association studies to provide insights into gene-trait associations, multi-marker analysis of genomic annotation to perform gene-based analysis, genome-wide complex trait analysis-multivariate set-based association test-combo to determine gene prioritization, and polygenic priority scores to prioritize the causal genes to screen candidate genes. Subsequent Mendelian randomization analysis was performed to infer causality between the candidate genes and GDM and phenome-wide association study (PheWAS) analysis was used to explore the associations between selected genes and other characteristics. Furthermore, to gain a deeper understanding of the functional implications of these susceptibility genes, GeneMANIA analysis was used to determine the fundamental biological functions of the therapeutic targets and protein-protein interaction network analysis was performed to identify intracellular protein interactions. RESULTS We identified two novel susceptibility genes associated with GDM: NPC1 and KIAA1191. Magnetic resonance imaging revealed a strong correlation between NPC1 expression levels and a lower incidence of GDM (odds ratio: 0.922, 95% confidence interval: 0.866-0.981, p = 0.011). PheWAS at the gene level indicated that NPC1 was not associated with any other trait. The biological significance of this gene was evidenced by its strong association with sterol metabolism. CONCLUSION Our study identified NPC1 as a novel gene whose predicted expression level is linked to a reduced risk of GDM, providing new insights into the genetic framework of this disease.
Collapse
Affiliation(s)
- Yuping Shan
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hong Hu
- Clinical medicine, Nantong University, Nantong, China
| | - Anning Yang
- Department of Obstetrics and Gynecology, Qingdao Eighth People's Hospital, Qingdao, China
| | - Wendi Zhao
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yijing Chu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Bajinka O, Ouedraogo SY, Li N, Zhan X. Big data for neuroscience in the context of predictive, preventive, and personalized medicine. EPMA J 2025; 16:17-35. [PMID: 39991094 PMCID: PMC11842698 DOI: 10.1007/s13167-024-00393-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/28/2024] [Accepted: 12/11/2024] [Indexed: 02/25/2025]
Abstract
Accurate and precise diagnosis made the medicine the hallmark of evidence-based medicine. While attaining absolute patient satisfaction may seem impossible in the aspect of disease recurrent, personalized their mecidal conditions to their responsive treatment approach may save the day. The last generation approaches in medicine require advanced technologies that will lead to evidence-based medicine. One of the trending fields in this is the use of big data in predictive, preventive, and personalized medicine (3PM). This review dwelled through the practical examples in which big data tools harness neuroscience to add more individualized apporahes to the medical conditions in a bid to confer a more personalized treatment strategies. Moreover, the known breakthroughs of big data in 3PM, big data and 3PM in neuroscience, AI and neuroscience, limitations of big data with 3PM in neuroscience, and the challenges are thoroughly discussed. Finally, the prospects of incorporating big data in 3PM are as well discussed. The review could point out that the implications of big data in 3PM are still in their infancy and will require a holistic approach. While there is a need to carefully sensitize the community, convincing them will come under interdisciplinary and, to some extent, inter-professional collaborations, capacity building for professionals, and optimal coordination of the joint systems.
Collapse
Affiliation(s)
- Ousman Bajinka
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Serge Yannick Ouedraogo
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
3
|
Mitchell J, Camacho N, Shea P, Stopsack KH, Joseph V, Burren OS, Dhindsa RS, Nag A, Berchuck JE, O'Neill A, Abbasi A, Zoghbi AW, Alegre-Díaz J, Kuri-Morales P, Berumen J, Tapia-Conyer R, Emberson J, Torres JM, Collins R, Wang Q, Goldstein D, Matakidou A, Haefliger C, Anderson-Dring L, March R, Jobanputra V, Dougherty B, Carss K, Petrovski S, Kantoff PW, Offit K, Mucci LA, Pomerantz M, Fabre MA. Assessing the contribution of rare protein-coding germline variants to prostate cancer risk and severity in 37,184 cases. Nat Commun 2025; 16:1779. [PMID: 39971927 PMCID: PMC11839991 DOI: 10.1038/s41467-025-56944-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/10/2024] [Accepted: 02/05/2025] [Indexed: 02/21/2025] Open
Abstract
To assess the contribution of rare coding germline genetic variants to prostate cancer risk and severity, we perform here a meta-analysis of 37,184 prostate cancer cases and 331,329 male controls from five cohorts with germline whole exome or genome sequencing data, and one cohort with imputed array data. At the gene level, our case-control collapsing analysis confirms associations between rare damaging variants in four genes and increased prostate cancer risk: SAMHD1, BRCA2 and ATM at the study-wide significance level (P < 1×10-8), and CHEK2 at the suggestive threshold (P < 2.6×10-6). Our case-only analysis, reveals that rare damaging variants in AOX1 are associated with more aggressive disease (OR = 2.60 [1.75-3.83], P = 1.35×10-6), as well as confirming the role of BRCA2 in determining disease severity. At the single-variant level, our study reveals that a rare missense variant in TERT is associated with substantially reduced prostate cancer risk (OR = 0.13 [0.07-0.25], P = 4.67×10-10), and confirms rare non-synonymous variants in a further three genes associated with reduced risk (ANO7, SPDL1, AR) and in three with increased risk (HOXB13, CHEK2, BIK). Altogether, this work provides deeper insights into the genetic architecture and biological basis of prostate cancer risk and severity, with potential implications for clinical risk prediction and therapeutic strategies.
Collapse
Affiliation(s)
- Jonathan Mitchell
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| | - Niedzica Camacho
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Patrick Shea
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Konrad H Stopsack
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Vijai Joseph
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Oliver S Burren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ryan S Dhindsa
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Abhishek Nag
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - Amanda O'Neill
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ali Abbasi
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Anthony W Zoghbi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jesus Alegre-Díaz
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, Ciudad de México, Mexico
| | - Pablo Kuri-Morales
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, Ciudad de México, Mexico
- Instituto Tecnológico y de Estudios Superiores de Monterrey, Tecnológico, Monterrey, Nuevo León, Mexico
| | - Jaime Berumen
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, Ciudad de México, Mexico
| | - Roberto Tapia-Conyer
- Faculty of Medicine, National Autonomous University of Mexico, Copilco Universidad, Coyoacán, Ciudad de México, Mexico
| | - Jonathan Emberson
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Jason M Torres
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Rory Collins
- Clinical Trial Service Unit & Epidemiological Studies Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Quanli Wang
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| | - David Goldstein
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
| | - Athena Matakidou
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Carolina Haefliger
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Lauren Anderson-Dring
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ruth March
- Precision Medicine and Biosamples, R&D Oncology, AstraZeneca, Dublin, Ireland
| | - Vaidehi Jobanputra
- Institute for Genomic Medicine, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | | | - Keren Carss
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Philip W Kantoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Convergent Therapeutics, Cambridge, MA, USA
| | - Kenneth Offit
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- American Cancer Society, Atlanta, GA, USA
| | | | - Margarete A Fabre
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
4
|
Ito G, Ota Y, Yamaguchi K, Furukawa Y, Mochizuki S, Ahiko Y, Shida D. Genetic analysis for diagnosing local recurrence of sigmoid colon cancer mimicking a small intestinal tumor: a case report. World J Surg Oncol 2025; 23:57. [PMID: 39966961 PMCID: PMC11834688 DOI: 10.1186/s12957-025-03706-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/20/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND With recent advances in genetics research, genetic analysis is increasingly being used in clinical practice. We report a case in which genetic analysis aided in diagnosing a local recurrence of sigmoid colon cancer, initially suspected to be a primary neoplasm of the small intestine. CASE PRESENTATION A 61-year-old male underwent laparoscopic sigmoidectomy for stage IIIB sigmoid colon cancer, followed by 8 cycles of CAPOX adjuvant chemotherapy, one and a half years prior. A follow-up CT scan performed one and a half years postoperatively revealed a mass in the small intestine near the ileal end, adjacent to the staple line of the previous colonic anastomosis. PET imaging showed high uptake in the small intestine but no significant uptake at the site of the prior anastomotic ring. Based on these findings, a primary small intestine neoplasm was suspected, rather than a local recurrence of the sigmoid cancer, prompting laparoscopic surgery. Intraoperative findings revealed an inflamed mass in the ileum, approximately 30 cm proximal to the cecum, involving staples from the previous anastomotic site. Consequently, an ileocecal resection combined with resection of the prior colonic anastomosis was performed. Macroscopically, the resected specimen revealed a 25-mm Type 2 tumor in the ileum extending into the previous anastomotic site of the large intestine, while the colonic mucosa remained intact. Histopathological examination identified a moderately differentiated tubular adenocarcinoma, consistent with the histology of the primary sigmoid cancer, raising the possibility of local recurrence. To analyze the origin of the ileal tumor, we performed whole-genome sequencing and subsequent PCR direct sequencing. As a result, identical mutations in two key driver genes (KRAS c.35G > A and PIK3CA c.1624G > A), as well as a mutation in a passenger gene (BBS9 c.2218_2222del), were identified in the primary and ileal tumors. These findings confirmed that the ileal tumor was a local recurrence rather than a new primary malignancy. CONCLUSIONS The present case highlights the practical application of genetic analysis in clinical practice, particularly when clinical diagnosis and histopathological findings are inconclusive or conflicting.
Collapse
Affiliation(s)
- Go Ito
- Division of Frontier Surgery, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 1088639, Japan
| | - Yasunori Ota
- Department of Pathology, The Institute of Medical Science Research Hospital, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 1088639, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 1088639, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 1088639, Japan
| | - Satoshi Mochizuki
- Tokyo Gut Clinic, Gyoshokai Medical Corporation, 2F Ueno Bldg, 2-6-2, Kajicho, Chiyoda- ku, Tokyo, 1010044, Japan
| | - Yuka Ahiko
- Division of Frontier Surgery, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 1088639, Japan
| | - Dai Shida
- Division of Frontier Surgery, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 1088639, Japan.
| |
Collapse
|
5
|
Wei J, Resztak JA, Ranjbaran A, Alazizi A, Mair-Meijers HE, Slatcher RB, Zilioli S, Wen X, Luca F, Pique-Regi R. Functional characterization of eQTLs and asthma risk loci with scATAC-seq across immune cell types and contexts. Am J Hum Genet 2025; 112:301-317. [PMID: 39814021 DOI: 10.1016/j.ajhg.2024.12.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/27/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025] Open
Abstract
cis-regulatory elements (CREs) control gene transcription dynamics across cell types and in response to the environment. In asthma, multiple immune cell types play an important role in the inflammatory process. Genetic variants in CREs can also affect gene expression response dynamics and contribute to asthma risk. However, the regulatory mechanisms underlying control of transcriptional dynamics across different environmental contexts and cell types at single-cell resolution remain to be elucidated. To resolve this question, we performed single-cell ATAC-seq (scATAC-seq) in peripheral blood mononuclear cells (PBMCs) from 16 children with asthma. PBMCs were activated with phytohemagglutinin (PHA) or lipopolysaccharide (LPS) and treated with dexamethasone (DEX), an anti-inflammatory glucocorticoid. We analyzed changes in chromatin accessibility, measured transcription factor motif activity, and identified treatment- and cell-type-specific transcription factors that drive changes in both gene expression mean and variability. We observed a strong positive linear dependence between motif response and their target gene expression changes but a negative relationship with changes in target gene expression variability. This result suggests that an increase of transcription factor binding tightens the variability of gene expression around the mean. We then annotated genetic variants in chromatin accessibility peaks and response motifs, followed by computational fine-mapping of expression quantitative trait loci (eQTL) from a pediatric asthma cohort. We found that eQTLs were 5-fold enriched in peaks with response motifs and refined the credible set for 410 asthma risk genes, with 191 having the causal variant in response motifs. In conclusion, scATAC-seq enhances the understanding of molecular mechanisms for asthma risk variants mediated by gene expression.
Collapse
Affiliation(s)
- Julong Wei
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Justyna A Resztak
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Ali Ranjbaran
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | - Adnan Alazizi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA
| | | | | | - Samuele Zilioli
- Department of Psychology, Wayne State University, Detroit, MI, USA; Department of Family Medicine and Public Health Sciences, Wayne State University, Detroit, MI, USA
| | - Xiaoquan Wen
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francesca Luca
- Department of Human Genetics, University of Chicago, Chicago, IL 60637, USA.
| | - Roger Pique-Regi
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA; Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
6
|
Sahu S, Galloux M, Southon E, Caylor D, Sullivan T, Arnaudi M, Zanti M, Geh J, Chari R, Michailidou K, Papaleo E, Sharan SK. Saturation genome editing-based clinical classification of BRCA2 variants. Nature 2025; 638:538-545. [PMID: 39779848 DOI: 10.1038/s41586-024-08349-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/30/2023] [Accepted: 11/05/2024] [Indexed: 01/11/2025]
Abstract
Sequencing-based genetic tests have uncovered a vast array of BRCA2 sequence variants1. Owing to limited clinical, familial and epidemiological data, thousands of variants are considered to be variants of uncertain significance2-4 (VUS). Here we have utilized CRISPR-Cas9-based saturation genome editing in a humanized mouse embryonic stem cell line to determine the functional effect of VUS. We have categorized nearly all possible single nucleotide variants (SNVs) in the region that encodes the carboxylate-terminal DNA-binding domain of BRCA2. We have generated function scores for 6,551 SNVs, covering 96.4% of possible SNVs in exons 15-26 spanning BRCA2 residues 2479-3216. These variants include 1,282 SNVs that are categorized as missense VUS in the clinical variant database ClinVar, with 77.2% of these classified as benign and 20.4% classified as pathogenic using our functional score. Our assay provides evidence that 3,384 of the SNVs in the region are benign and 776 are pathogenic. Our classification aligns closely with pathogenicity data from ClinVar, orthogonal functional assays and computational meta predictors. We have integrated our embryonic stem cell-based BRCA2-saturation genome editing dataset with other available evidence and utilized the American College of Medical Genetics and Genomics/Association for Molecular Pathology guidelines for clinical classification of all possible SNVs. This classification is available as a sequence-function map and serves as a valuable resource for interpreting unidentified variants in the population and for physicians and genetic counsellors to assess BRCA2 VUS in patients.
Collapse
Affiliation(s)
- Sounak Sahu
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | | | - Eileen Southon
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Dylan Caylor
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Teresa Sullivan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Matteo Arnaudi
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Lyngby, Denmark
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| | - Maria Zanti
- Biostatistics Unit, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Josephine Geh
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA
| | - Raj Chari
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Kyriaki Michailidou
- Biostatistics Unit, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Elena Papaleo
- Cancer Systems Biology, Section for Bioinformatics, Department of Health and Technology, Technical University of Denmark, Lyngby, Denmark
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| | - Shyam K Sharan
- Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA.
| |
Collapse
|
7
|
Zeng Y, Feng C, Jiang Z, Du W, He S, Li X, Fan Y, Ouyang X, Huang B, Su Y, Wang S, Wei R, Dai Z, Jin P, Liu J, Yang Q. Genome-wide association studies with prolapsed gland of the third eyelid in dogs. Front Vet Sci 2025; 11:1520155. [PMID: 39926593 PMCID: PMC11804112 DOI: 10.3389/fvets.2024.1520155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/30/2024] [Accepted: 12/30/2024] [Indexed: 02/11/2025] Open
Abstract
Cherry eye, the common name for the prolapse of the third eyelid gland in dogs, is a widespread ophthalmic disease affecting dogs of various breeds. This condition severely affects the quality of life of affected dogs, and its underlying cause remains unresolved. In this study, 170K SNP microarray data were collected from 653 brachycephalic dogs and 788 brachycephalic and mesocephalic dogs. These two datasets were analyzed separately in genome-wide association studies (GWAS) involving 12 dog breeds affected by cherry eye. The GWAS analysis of 653 short-headed dogs revealed that four SNPs in the CFA3:15627075-15983629 bp region exceeded the genome-level significance threshold. Association analysis of this region also indicated that these four SNPs were strongly associated. Gene annotation showed that the region contained genes such as KIAA0825, FAM172A, and NR2F1, of which NR2F1 was associated with eye development. The results showed that GWAS analysis performed on 788 short- and medium-headed dogs identified five SNPs in the CFA22:15627075-15983629 bp region that exceeded the genome-level significance threshold, and association analysis was performed in this region, which showed that these five SNPs were strongly associated. In addition, 104 annotated genes were identified in both GWAS. To explore the genes involved in cherry eyes, we performed GO functional enrichment analysis. The genes involved in the high pathway were DIO3 and TTC8. In addition, an in-depth analysis revealed 33 genes associated with eye development and diseases. Our study provides new perspectives for further understanding cherry eye in dogs.
Collapse
Affiliation(s)
- Yu Zeng
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Cundong Feng
- School of Tropical Agriculture and Forestry (School of Agricultural and Rural Affairs, School of Rural Revitalization), Hainan University, Haikou, China
| | - Zheli Jiang
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Weian Du
- School of Stomatology and Medicine, Foshan University, Foshan, China
| | - Shan He
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Xingnuan Li
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Yi Fan
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Xiao Ouyang
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Bixin Huang
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Yan Su
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Siyu Wang
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Rongxing Wei
- Nanchang Police Dog Base of the Ministry of Public Security of China, Nanchang, China
| | - Zonghao Dai
- Nanchang Police Dog Base of the Ministry of Public Security of China, Nanchang, China
| | - Peng Jin
- Nanchang Police Dog Base of the Ministry of Public Security of China, Nanchang, China
| | - Jianyun Liu
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| | - Qianyong Yang
- Jiujiang Key Laboratory of Rare Disease Research, Jiujiang University, Jiujiang, China
- Jiangxi Provincial Key Laboratory of Cell Precision Therapy, School of Basic Medical Sciences, Jiujiang University, Jiujiang, China
- Jiujiang Innovation Center of Biosensor Technology and Application, Jiujiang University, Jiujiang, China
| |
Collapse
|
8
|
Méndez-Vidal C, Bravo-Gil N, Pérez-Florido J, Marcos-Luque I, Fernández RM, Fernández-Rueda JL, González-Del Pozo M, Martín-Sánchez M, Fernández-Suárez E, Mena M, Carmona R, Dopazo J, Borrego S, Antiñolo G. A genomic strategy for precision medicine in rare diseases: integrating customized algorithms into clinical practice. J Transl Med 2025; 23:86. [PMID: 39833864 PMCID: PMC11748347 DOI: 10.1186/s12967-025-06069-2] [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] [Academic Contribution Register] [Received: 07/04/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Despite the use of Next-Generation Sequencing (NGS) as the gold standard for the diagnosis of rare diseases, its clinical implementation has been challenging, limiting the cost-effectiveness of NGS and the understanding, control and safety essential for decision-making in clinical applications. Here, we describe a personalized NGS-based strategy integrating precision medicine into a public healthcare system and its implementation in the routine diagnosis process during a five-year pilot program. METHODS Our approach involved customized probe designs, the generation of virtual panels and the development of a personalized medicine module (PMM) for variant prioritization. This strategy was applied to 6500 individuals including 6267 index patients and 233 NGS-based carrier screenings. RESULTS Causative variants were identified in 2061 index patients (average 32.9%, ranging from 12 to 62% by condition). Also, 131 autosomal-recessive cases could be partially genetically diagnosed. These results led to over 5000 additional studies including carrier, prenatal and preimplantational tests or pharmacological and gene therapy treatments. CONCLUSION This strategy has shown promising improvements in the diagnostic rate, facilitating timely diagnosis and gradually expanding our services portfolio for rare diseases. The steps taken towards the integration of clinical and genomic data are opening new possibilities for conducting both retrospective and prospective healthcare studies. Overall, this study represents a major milestone in the ongoing efforts to improve our understanding and clinical management of rare diseases, a crucial area of medical research and care.
Collapse
Affiliation(s)
- Cristina Méndez-Vidal
- Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Nereida Bravo-Gil
- Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Javier Pérez-Florido
- Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
- Platform of Computational Medicine. Fundación Progreso y Salud (FPS). CDCA, University Hospital Virgen del Rocio, Seville, Spain
| | - Irene Marcos-Luque
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
| | - Raquel M Fernández
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
| | - José Luis Fernández-Rueda
- Platform of Computational Medicine. Fundación Progreso y Salud (FPS). CDCA, University Hospital Virgen del Rocio, Seville, Spain
| | - María González-Del Pozo
- Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Marta Martín-Sánchez
- Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Elena Fernández-Suárez
- Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Marcela Mena
- Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
| | - Rosario Carmona
- Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
- Platform of Computational Medicine. Fundación Progreso y Salud (FPS). CDCA, University Hospital Virgen del Rocio, Seville, Spain
| | - Joaquín Dopazo
- Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain
- Platform of Computational Medicine. Fundación Progreso y Salud (FPS). CDCA, University Hospital Virgen del Rocio, Seville, Spain
| | - Salud Borrego
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain.
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain.
| | - Guillermo Antiñolo
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Seville, Spain.
- Department of Maternofetal Medicine, Genetics and Reproduction, Institute of Biomedicine of Seville, IBiS/University Hospital Virgen del Rocio, CSIC/University of Seville, Seville, Spain.
| |
Collapse
|
9
|
Ooka T. The Era of Preemptive Medicine: Developing Medical Digital Twins through Omics, IoT, and AI Integration. JMA J 2025; 8:1-10. [PMID: 39926086 PMCID: PMC11799569 DOI: 10.31662/jmaj.2024-0213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 02/11/2025] Open
Abstract
Preemptive medicine represents a paradigm shift from reactive treatment to proactive disease prevention. The integration of omics technologies, the Internet of Things (IoT), and artificial intelligence (AI) has facilitated the development of personalized, predictive, and preemptive healthcare strategies. Omic technologies, such as genomics, proteomics, and metabolomics, provide comprehensive insights into molecular profile of an individual, revealing potential disease predispositions and health trajectories. IoT devices, such as wearables and smartphones, enable continuous and periodic monitoring of physiological parameters, thus providing a dynamic view of an individual's health status. AI algorithms analyze comprehensive and complex data from omics and IoT technologies to identify patterns and correlations that inform predictive models of disease risk, progression, and response to interventions. Medical digital twins, or virtual replicas of an individual's biological processes, have emerged as the cornerstone of preemptive medicine. The integration of omics, IoT, and AI enables the development of medical digital twins, which in turn allows for precise simulation of human physiological profiles, prediction of future health outcomes, and virtual individual clinical trials, facilitating personalized proactive interventions and preemptive disease control. This review demonstrates the convergence of omics, IoT, and AI in preemptive medicine, highlighting their potential to revolutionize healthcare by enabling early disease detection, personalized treatment strategies, and chronic disease prevention. We show how AI leverages omics and IoT in preemptive medicine through several case studies while also discussing the necessary data for developing medical digital twins and addressing ethical and social aspects that warrant consideration. Medical digital twins signify a fundamental transformation in health management, shifting from treating diseases after their occurrence to controlling them before their occurrence. This approach enhances the effectiveness of medical interventions and improves overall health outcomes, preparing for a healthier future.
Collapse
Affiliation(s)
- Tadao Ooka
- Department of Health Sciences, University of Yamanashi, Chuo, Japan
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
10
|
Hollingsworth EW, Liu TA, Alcantara JA, Chen CX, Jacinto SH, Kvon EZ. Rapid and quantitative functional interrogation of human enhancer variant activity in live mice. Nat Commun 2025; 16:409. [PMID: 39762235 PMCID: PMC11704014 DOI: 10.1038/s41467-024-55500-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/26/2023] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Functional analysis of non-coding variants associated with congenital disorders remains challenging due to the lack of efficient in vivo models. Here we introduce dual-enSERT, a robust Cas9-based two-color fluorescent reporter system which enables rapid, quantitative comparison of enhancer allele activities in live mice in less than two weeks. We use this technology to examine and measure the gain- and loss-of-function effects of enhancer variants previously linked to limb polydactyly, autism spectrum disorder, and craniofacial malformation. By combining dual-enSERT with single-cell transcriptomics, we characterise gene expression in cells where the enhancer is normally and ectopically active, revealing candidate pathways that may lead to enhancer misregulation. Finally, we demonstrate the widespread utility of dual-enSERT by testing the effects of fifteen previously uncharacterised rare and common non-coding variants linked to neurodevelopmental disorders. In doing so we identify variants that reproducibly alter the in vivo activity of OTX2 and MIR9-2 brain enhancers, implicating them in autism. Dual-enSERT thus allows researchers to go from identifying candidate enhancer variants to analysis of comparative enhancer activity in live embryos in under two weeks.
Collapse
Affiliation(s)
- Ethan W Hollingsworth
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Medical Scientist Training Program, University of California, Irvine School of Medicine, Irvine, CA, USA
| | - Taryn A Liu
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Joshua A Alcantara
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Cindy X Chen
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Sandra H Jacinto
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Evgeny Z Kvon
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA.
| |
Collapse
|
11
|
Van Der Kelen A, Li Piani L, Mertens J, Regin M, Couvreu de Deckersberg E, Van de Velde H, Sermon K, Tournaye H, Verpoest W, Hes FJ, Blockeel C, Spits C. The interplay between mitochondrial DNA genotypes, female infertility, ovarian response, and mutagenesis in oocytes. Hum Reprod Open 2024; 2025:hoae074. [PMID: 39830711 PMCID: PMC11739621 DOI: 10.1093/hropen/hoae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/11/2024] [Revised: 11/06/2024] [Indexed: 01/22/2025] Open
Abstract
STUDY QUESTION Is there an association between different mitochondrial DNA (mtDNA) genotypes and female infertility or ovarian response, and is the appearance of variants in the oocytes favored by medically assisted reproduction (MAR) techniques? SUMMARY ANSWER Ovarian response was negatively associated with global non-synonymous protein-coding homoplasmic variants but positively associated with haplogroup K; the number of oocytes retrieved in a cycle correlates with the number of heteroplasmic variants in the oocytes, principally with variants located in the hypervariable (HV) region and rRNA loci, as well as non-synonymous protein-coding variants. WHAT IS KNOWN ALREADY Several genes have been shown to be positively associated with infertility, and there is growing concern that MAR may facilitate the transmission of these harmful variants to offspring, thereby passing on infertility. The potential role of mtDNA variants in these two perspectives remains poorly understood. STUDY DESIGN SIZE DURATION This cohort study included 261 oocytes from 132 women (mean age: 32 ± 4 years) undergoing ovarian stimulation between 2019 and 2020 at an academic center. The oocyte mtDNA genotypes were examined for associations with the women's fertility characteristics. PARTICIPANTS/MATERIALS SETTING METHODS The mtDNA of the oocytes underwent deep sequencing, and the mtDNA genotypes were compared between infertile and fertile groups using Fisher's exact test. The impact of the mtDNA genotype on anti-Müllerian hormone (AMH) levels and the number of (mature) oocytes retrieved was assessed using the Mann-Whitney U test for univariate analysis and logistic regression for multivariate analysis. Additionally, we examined the associations of oocyte maturation stage, infertility status, number of ovarian stimulation units, and number of oocytes retrieved with the type and load of heteroplasmic variants using univariate analysis and Poisson or linear regression analysis. MAIN RESULTS AND THE ROLE OF CHANCE Neither homoplasmic mtDNA variants nor haplogroups in the oocytes were associated with infertility status or with AMH levels. Conversely, when the relationship between the number of oocytes retrieved and different mtDNA genotypes was examined, a positive association was observed between the number of metaphase (MII) oocytes (P = 0.005) and haplogroup K. Furthermore, the presence of global non-synonymous homoplasmic variants in the protein-coding region was significantly associated with a reduced number of total oocytes and MII oocytes retrieved (P < 0.001 for both). Regarding the type and load of heteroplasmic variants in the different regions, there were no significant associations according to maturation stage of the oocyte or to fertility status; however, the number of oocytes retrieved correlated positively with the total number of heteroplasmic variants, and specifically with non-synonymous protein-coding, HV and rRNA variants (P < 0.001 for all). LIMITATIONS REASONS FOR CAUTION The current work is constrained by its retrospective design and single-center approach, potentially limiting the generalizability of our findings. The small sample size for specific types of infertility restricts this aspect of the findings. WIDER IMPLICATIONS OF THE FINDINGS This work suggests that mitochondrial genetics may have an impact on ovarian response and corroborates previous findings indicating that the size of the oocyte cohort after stimulation correlates with the presence of potentially deleterious variants in the oocyte. Future epidemiological and functional studies based on the results of the current study will provide valuable insights to address gaps in knowledge to assess any prospective risks for MAR-conceived offspring. STUDY FUNDING/COMPETING INTERESTS This work was supported by the Research Foundation Flanders (FWO, Grant numbers 1506617N and 1506717N to C.S.), by the Fonds Wetenschappelijk Fonds, Willy Gepts Research Foundation of Universitair Ziekenhuis Brussel (Grant numbers WFWG14-15, WFWG16-43, and WFWG19-19 to C.S.), and by the Methusalem Grant of the Vrije Universiteit Brussel (to K.S.). M.R. and E.C.d.D. were supported predoctoral fellowships by the FWO, Grant numbers 1133622N and 1S73521N, respectively. The authors declare no conflict of interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Annelore Van Der Kelen
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
- Universitair Ziekenhuis Brussel (UZ Brussel), Brussels Health Campus, Centre for Medical Genetics, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Letizia Li Piani
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
- Universitair Ziekenhuis Brussel (UZ Brussel), Brussels Health Campus, Centre for Reproductive Medicine, Brussels IVF, Laarbeeklaan 101, 1090 Brussels, Belgium
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Infertility Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Joke Mertens
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Marius Regin
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Edouard Couvreu de Deckersberg
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Hilde Van de Velde
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
- Universitair Ziekenhuis Brussel (UZ Brussel), Brussels Health Campus, Centre for Reproductive Medicine, Brussels IVF, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Karen Sermon
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Herman Tournaye
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
- Universitair Ziekenhuis Brussel (UZ Brussel), Brussels Health Campus, Centre for Reproductive Medicine, Brussels IVF, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Willem Verpoest
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
- Universitair Ziekenhuis Brussel (UZ Brussel), Brussels Health Campus, Centre for Reproductive Medicine, Brussels IVF, Laarbeeklaan 101, 1090 Brussels, Belgium
- Department of Reproductive Medicine, Utrecht University Medical Centre, Utrecht, The Netherlands
| | - Frederik Jan Hes
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
- Universitair Ziekenhuis Brussel (UZ Brussel), Brussels Health Campus, Centre for Medical Genetics, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Christophe Blockeel
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
- Universitair Ziekenhuis Brussel (UZ Brussel), Brussels Health Campus, Centre for Reproductive Medicine, Brussels IVF, Laarbeeklaan 101, 1090 Brussels, Belgium
| | - Claudia Spits
- Vrije Universiteit Brussel (VUB), Brussels Health Campus/Faculty of Medicine and Pharmacy, Research Group Genetics, Reproduction and Development, Laarbeeklaan 103, 1090 Brussels, Belgium
| |
Collapse
|
12
|
Qi X, Ullah A, Yu W, Jin X, Liu H. Estimating the Genetic Risk of First-Degree Relatives for Chronic Diseases Using the Short Tandem Repeat Score as Model of Polygenic Inheritance. Biochem Genet 2024:10.1007/s10528-024-11003-0. [PMID: 39733222 DOI: 10.1007/s10528-024-11003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/08/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024]
Abstract
This study aims to establish a genetic risk assessment model based on a score of short tandem repeats (STRs) of polygenic inheritance. A total of 396 children and their biological parents were collected for STR genotyping. The numbers of tandem repeats of two alleles in one STR locus were assumed to be a quantitative genetic strength for disease incidence. The sums of 19 STR loci were considered a quantitative genetic strength per individual. Various thresholds of the STRs between paternal, maternal, and childhood data were recorded. As an exemplar, for thresholds of 25%, the first quarter = 1. All other samples = 0. The consistency rate for heredity (CH) was calculated from the difference in the morbidity of children between parents with and without disease groups. The ratio of observed CH to expected CH was defined as the heredity index (HI). Actual Pedigree data (finger-crossing test) confirmed the accuracy of the STR score. The genetic risk of first-degree relatives could be estimated using easily acquired data (incidence in an unrelated population). Our findings can provide a polygenic genetic model for estimating the incidence and genetic risk of chronic disease in first-degree relatives.
Collapse
Affiliation(s)
- Xia Qi
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Anwar Ullah
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Weijian Yu
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Xiaojun Jin
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, People's Republic of China
| | - Hui Liu
- College of Medical Laboratory, Dalian Medical University, Dalian, 116044, People's Republic of China.
| |
Collapse
|
13
|
Liang Q, Sun Y, Li M, Li R, Nie L, Lin L, Yu X. Association and function analysis of genetic variants and the risk of gestational diabetes mellitus in a southern Chinese population. Front Endocrinol (Lausanne) 2024; 15:1476222. [PMID: 39777224 PMCID: PMC11703716 DOI: 10.3389/fendo.2024.1476222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/05/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Background Gestational diabetes mellitus (GDM) is a complex metabolic disease that has short-term and long-term adverse effects on mothers and infants. However, the specific pathogenic mechanism has not been elucidated. Objective The aim of this study was to confirm the associations between candidate genetic variants (rs4134819, rs720918, rs2034410, rs11109509, and rs12524768) and GDM risk and prediction in a southern Chinese population. Methods Candidate variants were genotyped in 538 GDM cases and 626 healthy controls. The odds ratio (OR) and its corresponding 95% confidence interval (CI) were calculated to assess the associations between genotypes and GDM risk. Then, the false-positive report probability (FPRP) analysis was adopted to confirm the significant associations, and bioinformatics tools were used to explore the potential biological function of studied variants. Finally, risk factors of genetic variants and clinical indicators identified by logistics regression were used to construct a nomogram model for GDM prediction. Results It was shown that the XAB2 gene rs4134819 was significantly associated with GDM susceptibility (CT vs. CC: adjusted OR = 1.38, 95% CI: 1.01-1.87, p = 0.044; CT/TT vs. CC: crude OR = 1.42, 95% CI: 1.08-1.86, p = 0.013). Functional analysis suggested that rs4134819 can alter the specific transcription factors (CPE bind and GATE-1) binding to the promoter of the XAB2 gene, regulating the transcription of XAB2. The nomogram established with factors such as age, FPG, HbA1c, 1hPG, 2hPG, TG, and rs4134819 showed a good discriminated and calibrated ability with an area under the curve (AUC) = 0.931 and a Hosmer-Lemeshow test p-value > 0.05. Conclusion The variant rs4134819 can significantly alter the susceptibility of the Chinese population to GDM possibly by regulating the transcription of functional genes. The nomogram prediction model constructed with genetic variants and clinical factors can help distinguish high-risk GDM individuals.
Collapse
Affiliation(s)
- Qiulian Liang
- School of Public Health and Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| | - Yan Sun
- School of Public Health and Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| | - Ming Li
- Department of Histology and Embryology, School of Basic Medicine, Hunan University of Medicine, Huaihua, China
| | - Ruiqi Li
- School of Public Health and Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| | - Lijie Nie
- School of Public Health and Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| | - Lin Lin
- The Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Xiangyuan Yu
- School of Public Health and Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, China
| |
Collapse
|
14
|
Spence JP, Mostafavi H, Ota M, Milind N, Gjorgjieva T, Smith CJ, Simons YB, Sella G, Pritchard JK. Specificity, length, and luck: How genes are prioritized by rare and common variant association studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.12.628073. [PMID: 39935885 PMCID: PMC11812597 DOI: 10.1101/2024.12.12.628073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 02/13/2025]
Abstract
Standard genome-wide association studies (GWAS) and rare variant burden tests are essential tools for identifying trait-relevant genes. Although these methods are conceptually similar, we show by analyzing association studies of 209 quantitative traits in the UK Biobank that they systematically prioritize different genes. This raises the question of how genes should ideally be prioritized. We propose two prioritization criteria: 1) trait importance - how much a gene quantitatively affects a trait; and 2) trait specificity - a gene's importance for the trait under study relative to its importance across all traits. We find that GWAS prioritize genes near trait-specific variants, while burden tests prioritize trait-specific genes. Because non-coding variants can be context specific, GWAS can prioritize highly pleiotropic genes, while burden tests generally cannot. Both study designs are also affected by distinct trait-irrelevant factors, complicating their interpretation. Our results illustrate that burden tests and GWAS reveal different aspects of trait biology and suggest ways to improve their interpretation and usage.
Collapse
Affiliation(s)
| | - Hakhamanesh Mostafavi
- Department of Genetics, Stanford University
- Center for Human Genetics and Genomics, New York University School of Medicine
- Department of Population Health, New York University School of Medicine
| | - Mineto Ota
- Department of Genetics, Stanford University
| | | | | | | | - Yuval B. Simons
- Department of Genetics, Stanford University
- Section of Genetic Medicine, University of Chicago
- Department of Human Genetics, University of Chicago
| | - Guy Sella
- Department of Biological Sciences, Columbia University
- Program for Mathematical Genomics, Columbia University
| | - Jonathan K. Pritchard
- Department of Genetics, Stanford University
- Department of Biology, Stanford University
| |
Collapse
|
15
|
Sadowski M, Thompson M, Mefford J, Haldar T, Oni-Orisan A, Border R, Pazokitoroudi A, Cai N, Ayroles JF, Sankararaman S, Dahl AW, Zaitlen N. Characterizing the genetic architecture of drug response using gene-context interaction methods. CELL GENOMICS 2024; 4:100722. [PMID: 39637863 DOI: 10.1016/j.xgen.2024.100722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/28/2024] [Revised: 06/24/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
Identifying factors that affect treatment response is a central objective of clinical research, yet the role of common genetic variation remains largely unknown. Here, we develop a framework to study the genetic architecture of response to commonly prescribed drugs in large biobanks. We quantify treatment response heritability for statins, metformin, warfarin, and methotrexate in the UK Biobank. We find that genetic variation modifies the primary effect of statins on LDL cholesterol (9% heritable) as well as their side effects on hemoglobin A1c and blood glucose (10% and 11% heritable, respectively). We identify dozens of genes that modify drug response, which we replicate in a retrospective pharmacogenomic study. Finally, we find that polygenic score (PGS) accuracy varies up to 2-fold depending on treatment status, showing that standard PGSs are likely to underperform in clinical contexts.
Collapse
Affiliation(s)
- Michal Sadowski
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA.
| | - Mike Thompson
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Joel Mefford
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tanushree Haldar
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA; Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Akinyemi Oni-Orisan
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA; Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA 94143, USA
| | - Richard Border
- Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computational Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ali Pazokitoroudi
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Na Cai
- Helmholtz Pioneer Campus, Helmholtz Munich, 85764 Neuherberg, Germany; Computational Health Centre, Helmholtz Munich, 85764 Neuherberg, Germany; School of Medicine and Health, Technical University of Munich, 80333 Munich, Germany
| | - Julien F Ayroles
- Department of Ecology and Evolution, Princeton University, Princeton, NJ 08544, USA; Lewis Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Sriram Sankararaman
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computer Science, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computational Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Andy W Dahl
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Noah Zaitlen
- Bioinformatics Interdepartmental Program, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Neurology, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Computational Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
16
|
Forero DA, Bonilla DA, González-Giraldo Y, Patrinos GP. An overview of key online resources for human genomics: a powerful and open toolbox for in silico research. Brief Funct Genomics 2024; 23:754-764. [PMID: 38993146 DOI: 10.1093/bfgp/elae029] [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] [Academic Contribution Register] [Received: 03/15/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
Recent advances in high-throughput molecular methods have led to an extraordinary volume of genomics data. Simultaneously, the progress in the computational implementation of novel algorithms has facilitated the creation of hundreds of freely available online tools for their advanced analyses. However, a general overview of the most commonly used tools for the in silico analysis of genomics data is still missing. In the current article, we present an overview of commonly used online resources for genomics research, including over 50 tools. This selection will be helpful for scientists with basic or intermediate skills in the in silico analyses of genomics data, such as researchers and students from wet labs seeking to strengthen their computational competencies. In addition, we discuss current needs and future perspectives within this field.
Collapse
Affiliation(s)
- Diego A Forero
- School of Health and Sport Sciences, Fundación Universitaria del Área Andina, Bogotá, Colombia
| | - Diego A Bonilla
- Research Division, Dynamical Business & Science Society - DBSS International SAS, Bogotá, Colombia
- Hologenomiks Research Group, Department of Genetics, Physical Anthropology and Animal Physiology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Yeimy González-Giraldo
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Individualized Therapy, Department of Pharmacy, School of Health Science, University of Patras, Patras, Greece
- Clinical Bioinformatics Unit, Department of Pathology, School of Medicine and Health Sciences, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al-AIn, Abu Dhabi, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al-AIn, Abu Dhabi, United Arab Emirates
| |
Collapse
|
17
|
Huang K, Zeng T, Koc S, Pettet A, Zhou J, Jain M, Sun D, Ruiz C, Ren H, Howe L, Richardson TG, Cortes A, Aiello K, Branson K, Pfenning A, Engreitz JM, Zhang MJ, Leskovec J. Small-cohort GWAS discovery with AI over massive functional genomics knowledge graph. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.03.24318375. [PMID: 39677475 PMCID: PMC11643201 DOI: 10.1101/2024.12.03.24318375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/17/2024]
Abstract
Genome-wide association studies (GWASs) have identified tens of thousands of disease associated variants and provided critical insights into developing effective treatments. However, limited sample sizes have hindered the discovery of variants for uncommon and rare diseases. Here, we introduce KGWAS, a novel geometric deep learning method that leverages a massive functional knowledge graph across variants and genes to improve detection power in small-cohort GWASs significantly. KGWAS assesses the strength of a variant's association to disease based on the aggregate GWAS evidence across molecular elements interacting with the variant within the knowledge graph. Comprehensive simulations and replication experiments showed that, for small sample sizes ( N =1-10K), KGWAS identified up to 100% more statistically significant associations than state-of-the-art GWAS methods and achieved the same statistical power with up to 2.67× fewer samples. We applied KGWAS to 554 uncommon UK Biobank diseases ( N case <5K) and identified 183 more associations (46.9% improvement) than the original GWAS, where the gain further increases to 79.8% for 141 rare diseases (N case <300). The KGWAS-only discoveries are supported by abundant functional evidence, such as rs2155219 (on 11q13) associated with ulcerative colitis potentially via regulating LRRC32 expression in CD4+ regulatory T cells, and rs7312765 (on 12q12) associated with the rare disease myasthenia gravis potentially via regulating PPHLN1 expression in neuron-related cell types. Furthermore, KGWAS consistently improves downstream analyses such as identifying disease-specific network links for interpreting GWAS variants, identifying disease-associated genes, and identifying disease-relevant cell populations. Overall, KGWAS is a flexible and powerful AI model that integrates growing functional genomics data to discover novel variants, genes, cells, and networks, especially valuable for small cohort diseases.
Collapse
|
18
|
Vanneste M, Hoskens H, Goovaerts S, Matthews H, Devine J, Aponte JD, Cole J, Shriver M, Marazita ML, Weinberg SM, Walsh S, Richmond S, Klein OD, Spritz RA, Peeters H, Hallgrímsson B, Claes P. Syndrome-informed phenotyping identifies a polygenic background for achondroplasia-like facial variation in the general population. Nat Commun 2024; 15:10458. [PMID: 39622794 PMCID: PMC11612227 DOI: 10.1038/s41467-024-54839-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/29/2023] [Accepted: 11/21/2024] [Indexed: 12/06/2024] Open
Abstract
Human craniofacial shape is highly variable yet highly heritable with numerous genetic variants interacting through multiple layers of development. Here, we hypothesize that Mendelian phenotypes represent the extremes of a phenotypic spectrum and, using achondroplasia as an example, we introduce a syndrome-informed phenotyping approach to identify genomic loci associated with achondroplasia-like facial variation in the general population. We compare three-dimensional facial scans from 43 individuals with achondroplasia and 8246 controls to calculate achondroplasia-like facial scores. Multivariate GWAS of the control scores reveals a polygenic basis for facial variation along an achondroplasia-specific shape axis, identifying genes primarily involved in skeletal development. Jointly modeling these genes in two independent control samples, both human and mouse, shows craniofacial effects approximating the characteristic achondroplasia phenotype. These findings suggest that both complex and Mendelian genetic variation act on the same developmentally determined axes of facial variation, providing insights into the genetic intersection of complex traits and Mendelian disorders.
Collapse
Affiliation(s)
| | - Hanne Hoskens
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Seppe Goovaerts
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
| | - Harold Matthews
- Department of Human Genetics, KU Leuven, Leuven, Belgium
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
| | - Jay Devine
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium
| | - Jose D Aponte
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Joanne Cole
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Mark Shriver
- Department of Anthropology, Pennsylvania State University, State College, PA, USA
| | - Mary L Marazita
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seth M Weinberg
- Center for Craniofacial and Dental Genetics, Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Susan Walsh
- Department of Biology, Indiana University Purdue University Indianapolis, Indianapolis, IN, USA
| | - Stephen Richmond
- Applied Clinical Research and Public Health, School of Dentistry, Cardiff University, Cardiff, UK
| | - Ophir D Klein
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Richard A Spritz
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Hilde Peeters
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
| | - Benedikt Hallgrímsson
- Department of Cell Biology & Anatomy, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
- McCaig Bone and Joint Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| | - Peter Claes
- Department of Human Genetics, KU Leuven, Leuven, Belgium.
- Medical Imaging Research Center, University Hospitals Leuven, Leuven, Belgium.
- Department of Electrical Engineering, ESAT/PSI, KU Leuven, Leuven, Belgium.
| |
Collapse
|
19
|
Elgedawy GA, Elabd NS, Salem RH, Awad SM, Amer AA, Torayah MM, El-Koa AA, Abozeid M, Montaser BA, Aboshabaan HS, Abdelkreem M, Helal ML. FURIN, IFNL4, and TLR2 gene polymorphisms in relation to COVID-19 severity: a case-control study in Egyptian patients. Infection 2024; 52:2213-2229. [PMID: 38703289 PMCID: PMC11621141 DOI: 10.1007/s15010-024-02266-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/01/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND AND AIM A wide range of clinical manifestations and outcomes, including liver injury, have been reported in COVID-19 patients. We investigated the association of three substantial gene polymorphisms (FURIN, IFNL4, and TLR2) with COVID-19 disease susceptibility and severity to help predict prognosis. METHODS 150 adult COVID-19-assured cases were categorized as follows: 78 patients with a non-severe presentation, 39 patients with severe disease, and 33 critically ill patients. In addition, 74 healthy controls were included. Clinical and laboratory evaluations were carried out, including complete and differential blood counts, D-dimer, lactate dehydrogenase (LDH), C-reactive protein (CRP), procalcitonin, ferritin, interleukin-6 (Il-6), and liver and kidney functions. FURIN (rs6226), IFNL4 (rs12979860), and TLR2 (rs3804099) genotyping allelic discrimination assays were conducted using real-time PCR. RESULTS The FURIN, IFNL4, and TLR2 genotypes and their alleles differed significantly between COVID-19 patients and controls, as well as between patients with severe or critical illness and those with a non-severe presentation. According to a multivariable regression analysis, FURIN (C/T + T/T) and TLR2 (T/C + C/C) mutants were associated with COVID-19 susceptibility, with odds ratios of 3.293 and 2.839, respectively. FURIN C/C and IFNL4 T/T mutants were significantly linked to severe and critical illnesses. Multivariate regression analysis showed that FURIN (G/C + C/C) genotypes and IFNL4 T/T homozygosity were independent risk factors associated with increased mortality. CONCLUSION FURIN, IFNL4, and TLR2 gene variants are associated with the risk of COVID-19 occurrence as well as increased severity and poor outcomes in Egyptian patients.
Collapse
Affiliation(s)
- Gamalat A Elgedawy
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Naglaa S Elabd
- Faculty of Medicine, Department of Tropical Medicine, Menoufia University, Shebin El-Kom, Menoufia, 32511, Egypt.
| | - Radwa H Salem
- Department of Clinical Microbiology and Immunology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Samah M Awad
- Department of Clinical Microbiology and Immunology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Amany A Amer
- Faculty of Medicine, Department of Tropical Medicine, Menoufia University, Shebin El-Kom, Menoufia, 32511, Egypt
| | - Mohammad M Torayah
- Faculty of Medicine, Department of Critical Care Medicine, Menoufia University, Shebin El-Kom, Egypt
| | - Amal A El-Koa
- Faculty of Medicine, Department of Chest Diseases and Tuberculosis, Menoufia University, Shebin El‑Kom, Menoufia, Egypt
| | - Mai Abozeid
- Department of Hepatology and Gastroenterology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, 32511, Egypt.
| | - Belal A Montaser
- Faculty of Medicine, Department of Clinical Pathology, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Hind S Aboshabaan
- Ph.D. of Biochemistry, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| | - Mervat Abdelkreem
- Department of Hepatology and Gastroenterology, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, 32511, Egypt
| | - Marwa L Helal
- Department of Clinical Biochemistry and Molecular Diagnostics, National Liver Institute, Menoufia University, Shebin El-Kom, Menoufia, Egypt
| |
Collapse
|
20
|
Belli O, Karava K, Farouni R, Platt RJ. Multimodal scanning of genetic variants with base and prime editing. Nat Biotechnol 2024:10.1038/s41587-024-02439-1. [PMID: 39533106 DOI: 10.1038/s41587-024-02439-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/21/2023] [Accepted: 09/18/2024] [Indexed: 11/16/2024]
Abstract
Mutational scanning connects genetic variants to phenotype, enabling the interrogation of protein functions, interactions and variant pathogenicity. However, current methodologies cannot efficiently engineer customizable sets of diverse genetic variants in endogenous loci across cellular contexts in high throughput. Here, we combine cytosine and adenine base editors and a prime editor to assess the pathogenicity of a broad spectrum of variants in the epithelial growth factor receptor gene (EGFR). Using pooled base editing and prime editing guide RNA libraries, we install tens of thousands of variants spanning the full coding sequence of EGFR in multiple cell lines and assess the role of these variants in tumorigenesis and resistance to tyrosine kinase inhibitors. Our EGFR variant scan identifies important hits, supporting the robustness of the approach and revealing underappreciated routes to EGFR activation and drug response. We anticipate that multimodal precision mutational scanning can be applied broadly to characterize genetic variation in any genetic element of interest at high and single-nucleotide resolution.
Collapse
Affiliation(s)
- Olivier Belli
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Kyriaki Karava
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Rick Farouni
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Randall J Platt
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
- Basel Research Centre for Child Health, Basel, Switzerland.
- Department of Chemistry, University of Basel, Basel, Switzerland.
- NCCR Molecular Systems Engineering, Basel, Switzerland.
| |
Collapse
|
21
|
Ferris LJ, Hornsey MJ, Morosoli JJ, Milfont TL, Barlow FK. A 30-nation investigation of lay heritability beliefs. PUBLIC UNDERSTANDING OF SCIENCE (BRISTOL, ENGLAND) 2024; 33:940-960. [PMID: 38664920 PMCID: PMC11528883 DOI: 10.1177/09636625241245030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 10/25/2024]
Abstract
Lay beliefs about human trait heritability are consequential for cooperation and social cohesion, yet there has been no global characterisation of these beliefs. Participants from 30 countries (N = 6128) reported heritability beliefs for intelligence, personality, body weight and criminality, and transnational factors that could influence these beliefs were explored using public nation-level data. Globally, mean lay beliefs differ from published heritability (h2) estimated by twin studies, with a worldwide majority overestimating the heritability of personality and intelligence, and underestimating body weight and criminality. Criminality was seen as substantially less attributable to genes than other traits. People from countries with high infant mortality tended to ascribe greater heritability for most traits, relative to people from low infant mortality countries. This study provides the first systematic foray into worldwide lay heritability beliefs. Future research must incorporate diverse global perspectives to further contextualise and extend upon these findings.
Collapse
Affiliation(s)
- Laura J. Ferris
- Laura J. Ferris, The University of Queensland, St Lucia, QLD 4072, Australia.
| | | | - José J. Morosoli
- University College London, UK; The University of Queensland, Australia; QIMR Berghofer Medical Research Institute, Australia
| | | | | |
Collapse
|
22
|
Chi P, Ou G, Liu S, Ma Q, Lu Y, Li J, Li J, Qi Q, Han Z, Zhang Z, Liu Q, Guo L, Chen J, Wang X, Huang W, Li L, Deng D. Cryo-EM structure of the human subcortical maternal complex and the associated discovery of infertility-associated variants. Nat Struct Mol Biol 2024; 31:1798-1807. [PMID: 39379527 DOI: 10.1038/s41594-024-01396-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/06/2023] [Accepted: 08/28/2024] [Indexed: 10/10/2024]
Abstract
The functionally conserved subcortical maternal complex (SCMC) is essential for early embryonic development in mammals. Reproductive disorders caused by pathogenic variants in NLRP5, TLE6 and OOEP, three core components of the SCMC, have attracted much attention over the past several years. Evaluating the pathogenicity of a missense variant in the SCMC is limited by the lack of information on its structure, although we recently solved the structure of the mouse SCMC and proposed that reproductive disorders caused by pathogenic variants are related to the destabilization of the SCMC core complex. Here we report the cryogenic electron microscopy structure of the human SCMC and uncover that the pyrin domain of NLRP5 is essential for the stability of SCMC. By combining prediction of SCMC stability and in vitro reconstitution, we provide a method for identifying deleterious variants, and we successfully identify a new pathogenic variant of TLE6 (p.A396T). Thus, on the basis of the structure of the human SCMC, we offer a strategy for the diagnosis of reproductive disorders and the discovery of new infertility-associated variants.
Collapse
Affiliation(s)
- Pengliang Chi
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Guojin Ou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- Clinical Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Sibei Liu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qianhong Ma
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yuechao Lu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jinhong Li
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jialu Li
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- NHC key Laboratory of Chronobiology, Sichuan University, Chengdu, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Qianqian Qi
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- Clinical Laboratory, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhuo Han
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- NHC key Laboratory of Chronobiology, Sichuan University, Chengdu, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Zihan Zhang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- NHC key Laboratory of Chronobiology, Sichuan University, Chengdu, China
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China
| | - Qingting Liu
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Guo
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Jing Chen
- Laboratory of Pediatric Surgery, Department of Pediatric Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Wang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Wei Huang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
- Department of Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Lei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Stem Cell and Regeneration, Beijing Institute of Stem Cell and Regenerative Medicine, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, China
| | - Dong Deng
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Disease of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
- NHC key Laboratory of Chronobiology, Sichuan University, Chengdu, China.
- Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, China.
| |
Collapse
|
23
|
Dewell SL, Muglia KA, Graves LY, Joseph R, Mangold KL, Roselli LG, Ersig AL, Walker TK. Essentials of genomics in nursing undergraduate education: A discussion paper. Nurse Educ Pract 2024; 81:104175. [PMID: 39481272 DOI: 10.1016/j.nepr.2024.104175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2024] [Revised: 10/14/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024]
Abstract
AIM To map the 2021 American Association of Colleges of Nursing Essentials to the American Nurses Association Essentials of Genomic Nursing for all nurses and provide resources for nursing faculty to support the seamless integration of genomics into existing undergraduate curricula. BACKGROUND Since the completion of the Human Genome Project in 2003, rapid advancements in genomic science leading to practical applications of genomics have revolutionized all areas of healthcare. Nursing is built on foundational life sciences, including genomics. As the largest segment of the healthcare workforce, who spend the most time with patients and families, nurses play a critical role in healthcare teams integrating genomic knowledge into patient care to improve health and well-being. Consequently, nurses must be equipped with foundational genomic knowledge and skills during their undergraduate education. However, there is wide variability in whether and how nursing programs have incorporated genomics into their curricula. Additionally, nursing faculty may have limited knowledge of foundational genomic concepts and lack confidence in teaching genomics. DESIGN Discussion paper METHODS: We aligned domains from the American Association of Colleges of Nursing Essentials and American Nurses Association Essentials of Genomic Nursing. RESULTS A map illustrating alignment in multiple areas, which provide examples of ways to integrate genomics into existing nursing curricula. CONCLUSION Although based on domains developed in the United States, the map, curricular resources, example learning outcomes, and clinical vignettes can be used by nursing faculty globally to prepare future nurses who are competent in providing genomics-informed nursing care on entry-to-practice.
Collapse
Affiliation(s)
- Sarah L Dewell
- Thompson Rivers University, School of Nursing, 805 TRU Way, Kamloops, BC V2C 0C8, Canada.
| | - Kathleen A Muglia
- Marquette University, College of Nursing, Clark Hall, 510 N. 16th St, Milwaukee, Wi 53233 & Child and Adolescent Psychiatry, Advocate Children's Hospital, 1775 Dempster St., Park Ridge, IL60068, USA.
| | - Letitia Y Graves
- School of Nursing, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-1132, USA.
| | - Rachel Joseph
- Liberty University, 1971 University Blvd., Lynchburg, VA 24515, USA.
| | - Kara L Mangold
- Mayo Clinic Center for Individualized Medicine, 13400 E. Shea Blvd., Scottsdale, AZ 85259, USA.
| | - Laura Grayson Roselli
- Biology Department, Rowan College at Burlington County, 900 College Circle Mount Laurel, NJ 08054, USA.
| | - Anne L Ersig
- University of Wisconsin-Madison School of Nursing, 701 Highland Ave, Madison, WI 53705, USA.
| | - Trina K Walker
- Creighton University, College of Nursing, 2500 California Plaza, Omaha, NE 68178, USA.
| |
Collapse
|
24
|
Lancaster MA. Unraveling mechanisms of human brain evolution. Cell 2024; 187:5838-5857. [PMID: 39423803 PMCID: PMC7617105 DOI: 10.1016/j.cell.2024.08.052] [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] [Academic Contribution Register] [Received: 02/15/2024] [Revised: 06/19/2024] [Accepted: 08/28/2024] [Indexed: 10/21/2024]
Abstract
Evolutionary changes in human brain structure and function have enabled our specialized cognitive abilities. How these changes have come about genetically and functionally has remained an open question. However, new methods are providing a wealth of information about the genetic, epigenetic, and transcriptomic differences that set the human brain apart. Combined with in vitro models that allow access to developing brain tissue and the cells of our closest living relatives, the puzzle pieces are now coming together to yield a much more complete picture of what is actually unique about the human brain. The challenge now will be linking these observations and making the jump from correlation to causation. However, elegant genetic manipulations are now possible and, when combined with model systems such as organoids, will uncover a mechanistic understanding of how evolutionary changes at the genetic level have led to key differences in development and function that enable human cognition.
Collapse
Affiliation(s)
- Madeline A Lancaster
- MRC Laboratory of Molecular Biology, Cambridge, UK; Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
25
|
Cunningham AG, Gorospe M. Striving for clarity in language about gene expression. Nucleic Acids Res 2024; 52:10747-10753. [PMID: 39271127 PMCID: PMC11472038 DOI: 10.1093/nar/gkae764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/10/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
What do we mean when we say 'gene expression'? In the decades following Crick's 1958 central dogma of molecular biology, whereby genetic information flows from DNA (genes) to RNA (transcripts) to protein (products), we have learned a great deal about DNA, RNA, proteins, and the ensuing phenotypic changes. With the advent of high-throughput technologies (1990s), molecular biologists and computer scientists forged critical collaborations to understand the vast amount of data being generated, rapidly escalating gene expression research to the 'omics' level: entire sets of genes (genomes), transcribed RNAs (transcriptomes), and synthesized proteins (proteomes). However, some concessions came to be made for molecular biologists and computer scientists to understand each other-one of the most prevalent being the increasingly widespread use of 'gene' to mean 'RNAs originating from a DNA segment'. This loosening of terminology, we will argue, creates ambiguity and confusion. We propose guidelines to increase precision and clarity when communicating about gene expression, most notably to reserve 'gene' for the DNA template and 'transcript' for the RNA transcribed from that gene. Striving to use perspicuous terminology will promote rigorous gene expression science and accelerate discovery in this highly promising area of biology.
Collapse
Affiliation(s)
- Ana S G Cunningham
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
26
|
Liu S, Liu Y, Gu Y, Lin X, Zhu H, Liu H, Xu Z, Cheng S, Lan X, Li L, Huang M, Li H, Nielsen R, Davies RW, Albrechtsen A, Chen GB, Qiu X, Jin X, Huang S. Utilizing non-invasive prenatal test sequencing data for human genetic investigation. CELL GENOMICS 2024; 4:100669. [PMID: 39389018 PMCID: PMC11602596 DOI: 10.1016/j.xgen.2024.100669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 12/23/2023] [Revised: 07/22/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
Non-invasive prenatal testing (NIPT) employs ultra-low-pass sequencing of maternal plasma cell-free DNA to detect fetal trisomy. Its global adoption has established NIPT as a large human genetic resource for exploring genetic variations and their associations with phenotypes. Here, we present methods for analyzing large-scale, low-depth NIPT data, including customized algorithms and software for genetic variant detection, genotype imputation, family relatedness, population structure inference, and genome-wide association analysis of maternal genomes. Our results demonstrate accurate allele frequency estimation and high genotype imputation accuracy (R2>0.84) for NIPT sequencing depths from 0.1× to 0.3×. We also achieve effective classification of duplicates and first-degree relatives, along with robust principal-component analysis. Additionally, we obtain an R2>0.81 for estimating genetic effect sizes across genotyping and sequencing platforms with adequate sample sizes. These methods offer a robust theoretical and practical foundation for utilizing NIPT data in medical genetic research.
Collapse
Affiliation(s)
- Siyang Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; Shenzhen Key Laboratory of Pathogenic Microbes and Biosafety, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; BGI-Shenzhen, Shenzhen 518083, Guangdong, China; Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China.
| | - Yanhong Liu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Yuqin Gu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | - Xingchen Lin
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China
| | | | - Hankui Liu
- BGI Genomics, BGI-Shenzhen, Shenzhen 518083, Guangdong, China
| | - Zhe Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Shiyao Cheng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Xianmei Lan
- BGI-Shenzhen, Shenzhen 518083, Guangdong, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linxuan Li
- BGI-Shenzhen, Shenzhen 518083, Guangdong, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingxi Huang
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Hao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Rasmus Nielsen
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Anders Albrechtsen
- Bioinformatics Centre, Department of Biology, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Guo-Bo Chen
- Center for Productive Medicine, Department of Genetic and Genomic Medicine, Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, Zhejiang, China
| | - Xiu Qiu
- Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; Provincial Clinical Research Center for Child Health, Guangzhou 510623, China; Department of Women's Health, Provincial Key Clinical Specialty of Woman and Child Health, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xin Jin
- BGI-Shenzhen, Shenzhen 518083, Guangdong, China; The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, Guangdong, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China.
| | - Shujia Huang
- BGI-Shenzhen, Shenzhen 518083, Guangdong, China; Division of Birth Cohort Study, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China.
| |
Collapse
|
27
|
Liu S, Yao J, Lin L, Lan X, Wu L, He X, Kong N, Li Y, Deng Y, Xie J, Zhu H, Wu X, Li Z, Xiong L, Wang Y, Ren J, Qiu X, Zhao W, Gao Y, Chen Y, Su F, Zhou Y, Rao W, Zhang J, Hou G, Huang L, Li L, Liu X, Nie C, Luo L, Zhao M, Liu Z, Chen F, Lin S, Zhao L, Fu Q, Jiang D, Yin Y, Xu X, Wang J, Yang H, Wang R, Niu J, Wei F, Jin X, Liu S. Genome-wide association study of maternal plasma metabolites during pregnancy. CELL GENOMICS 2024; 4:100657. [PMID: 39389015 PMCID: PMC11602615 DOI: 10.1016/j.xgen.2024.100657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 03/09/2023] [Revised: 01/05/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Metabolites are key indicators of health and therapeutic targets, but their genetic underpinnings during pregnancy-a critical period for human reproduction-are largely unexplored. Using genetic data from non-invasive prenatal testing, we performed a genome-wide association study on 84 metabolites, including 37 amino acids, 24 elements, 13 hormones, and 10 vitamins, involving 34,394 pregnant Chinese women, with sample sizes ranging from 6,394 to 13,392 for specific metabolites. We identified 53 metabolite-gene associations, 23 of which are novel. Significant differences in genetic effects between pregnant and non-pregnant women were observed for 16.7%-100% of these associations, indicating gene-environment interactions. Additionally, 50.94% of genetic associations exhibited pleiotropy among metabolites and between six metabolites and eight pregnancy phenotypes. Mendelian randomization revealed potential causal relationships between seven maternal metabolites and 15 human traits and diseases. These findings provide new insights into the genetic basis of maternal plasma metabolites during pregnancy.
Collapse
Affiliation(s)
| | - Jilong Yao
- Shenzhen Maternity & Child Healthcare Hospital, Shenzhen 518000, Guangdong, China
| | - Liang Lin
- BGI Genomics, Shenzhen 518083, China
| | - Xianmei Lan
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Linlin Wu
- Shenzhen Maternity & Child Healthcare Hospital, Shenzhen 518000, Guangdong, China; Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen 518000, Guangdong, China
| | - Xuelian He
- Genetic and Precision Medical Center, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Hubei, Wuhan, China
| | | | - Yan Li
- BGI Research, Shenzhen 518083, China
| | - Yuqing Deng
- Peking University Shenzhen Hospital, Shenzhen 518035, Guangdong, China
| | - Jiansheng Xie
- Shenzhen Maternity & Child Healthcare Hospital, Shenzhen 518000, Guangdong, China
| | | | - Xiaoxia Wu
- Shenzhen Maternity & Child Healthcare Hospital, Shenzhen 518000, Guangdong, China; Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen 518000, Guangdong, China; Department of Obstetrics, Shenzhen Maternity & Child Healthcare Hospital, The First School of Clinical Medicine, Southern Medical University, Shenzhen 518000, Guangdong China
| | - Zilong Li
- BGI Research, Shenzhen 518083, China
| | - Likuan Xiong
- Baoan Women's and Children's Hospital, Jinan University, Shenzhen 518133, Guangdong, China
| | - Yuan Wang
- BGI Genomics, Shenzhen 518083, China
| | - Jinghui Ren
- Shenzhen People's Hospital, 2nd Clinical Medical College of Jinan University, Shenzhen 518020, Guangdong, China
| | | | - Weihua Zhao
- Shenzhen Second People Hospital, Shenzhen 518035, Guangdong, China
| | - Ya Gao
- BGI Research, Shenzhen 518083, China
| | - Yuanqing Chen
- Nanshan Medical Group Headquarters of Shenzhen, Shenzhen 518000, Guangdong, China
| | | | - Yun Zhou
- Luohu People's Hospital of Shenzhen, Shenzhen 518001, Guangdong, China
| | | | - Jing Zhang
- Shenzhen Nanshan Maternity & Child Healthcare Hospital, Shenzhen 518000, Guangdong, China
| | | | - Liping Huang
- Shenzhen Baoan District Shajing People's Hospital, Shenzhen 518104, Guangdong, Chinas
| | - Linxuan Li
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinhong Liu
- Shenzhen Longhua District Central Hospital, Shenzhen 518110, Guangdong, China
| | - Chao Nie
- BGI Research, Shenzhen 518083, China
| | - Liqiong Luo
- The People's Hospital of Longhua-Shenzhen, Shenzhen 518109, Guangdong, China
| | - Mei Zhao
- BGI Genomics, Shenzhen 518083, China
| | - Zengyou Liu
- Shenzhen Nanshan People's Hospital, Shenzhen 518052, Guangdong, China
| | | | - Shengmou Lin
- The University of Hong Kong - Shenzhen Hospital, Shenzhen 518038, Guangdong, China
| | | | - Qingmei Fu
- Baoan People's Hospital of Shen Zhen, Shenzhen 518100, Guangdong, China
| | - Dan Jiang
- BGI Genomics, Shenzhen 518083, China
| | - Ye Yin
- BGI, Shenzhen 518083, China
| | - Xun Xu
- BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, China
| | | | - Huanming Yang
- BGI Research, Shenzhen 518083, China; Guangdong Provincial Academician Workstation of BGI Synthetic Genomics, Shenzhen, China
| | - Rong Wang
- BGI Genomics, Shenzhen 518083, China
| | - Jianmin Niu
- Shenzhen Maternity & Child Healthcare Hospital, Shenzhen 518000, Guangdong, China.
| | - Fengxiang Wei
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen 518172, Guangdong, China.
| | - Xin Jin
- BGI Research, Shenzhen 518083, China; The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China.
| | - Siqi Liu
- BGI Research, Shenzhen 518083, China; BGI Genomics, Shenzhen 518083, China.
| |
Collapse
|
28
|
Kang JH, Lee Y, Kim DJ, Kim JW, Cheon MJ, Lee BC. Polygenic risk and rare variant gene clustering enhance cancer risk stratification for breast and prostate cancers. Commun Biol 2024; 7:1289. [PMID: 39384879 PMCID: PMC11464688 DOI: 10.1038/s42003-024-06995-9] [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] [Academic Contribution Register] [Received: 09/25/2023] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
Polygenic risk score (PRS) and rare monogenic variant screening are valuable tools for predicting cancer risk and identifying individuals at high risk. Integrating both common and rare genetic variants is crucial for accurate risk assessment. However, estimating the impacts of rare variants on cancer and combining them with PRS remains challenging. Here, we analyze 454,711 exome sequencing and 487,409 array UK Biobank samples, focusing on breast and prostate cancers. We introduce an expanded PRS (EPRS) approach, yielding a systematic model for more effective risk stratification. By prioritizing and clustering genes with cancer-specific rare variants based on odds ratios and population-attributable fraction, we refine risk stratification by combining both monogenic and polygenic effects. Individuals in high-PRS groups with rare high-impact gene variants show up to 15- and 22-fold higher risk for breast and prostate cancers, respectively, compared to those in the intermediate-PRS groups without rare variants. Combined risk profiles vary across distinct rare variant clusters within the same PRS group for both cancers. Our EPRS approach enhances risk stratification for breast and prostate cancers, offering important insights for future research and potential applications to other cancer types.
Collapse
Affiliation(s)
- Joon Ho Kang
- R&D division Genoplan Korea Inc, Seoul, 06611, Republic of Korea
| | - Youngkee Lee
- R&D division Genoplan Korea Inc, Seoul, 06611, Republic of Korea
| | - Dong Jun Kim
- R&D division Genoplan Korea Inc, Seoul, 06611, Republic of Korea
| | - Ji-Woong Kim
- R&D division Genoplan Korea Inc, Seoul, 06611, Republic of Korea
| | - Myeong Jae Cheon
- R&D division Genoplan Korea Inc, Seoul, 06611, Republic of Korea
| | - Byung-Chul Lee
- R&D division Genoplan Korea Inc, Seoul, 06611, Republic of Korea.
| |
Collapse
|
29
|
Chong JX, Berger SI, Baxter S, Smith E, Xiao C, Calame DG, Hawley MH, Rivera-Munoz EA, DiTroia S, Bamshad MJ, Rehm HL. Considerations for reporting variants in novel candidate genes identified during clinical genomic testing. Genet Med 2024; 26:101199. [PMID: 38944749 PMCID: PMC11456385 DOI: 10.1016/j.gim.2024.101199] [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] [Academic Contribution Register] [Received: 02/09/2024] [Revised: 06/18/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024] Open
Abstract
Since the first novel gene discovery for a Mendelian condition was made via exome sequencing, the rapid increase in the number of genes known to underlie Mendelian conditions coupled with the adoption of exome (and more recently, genome) sequencing by diagnostic testing labs has changed the landscape of genomic testing for rare diseases. Specifically, many individuals suspected to have a Mendelian condition are now routinely offered clinical ES. This commonly results in a precise genetic diagnosis but frequently overlooks the identification of novel candidate genes. Such candidates are also less likely to be identified in the absence of large-scale gene discovery research programs. Accordingly, clinical laboratories have both the opportunity, and some might argue a responsibility, to contribute to novel gene discovery, which should, in turn, increase the diagnostic yield for many conditions. However, clinical diagnostic laboratories must necessarily balance priorities for throughput, turnaround time, cost efficiency, clinician preferences, and regulatory constraints and often do not have the infrastructure or resources to effectively participate in either clinical translational or basic genome science research efforts. For these and other reasons, many laboratories have historically refrained from broadly sharing potentially pathogenic variants in novel genes via networks such as Matchmaker Exchange, much less reporting such results to ordering providers. Efforts to report such results are further complicated by a lack of guidelines for clinical reporting and interpretation of variants in novel candidate genes. Nevertheless, there are myriad benefits for many stakeholders, including patients/families, clinicians, and researchers, if clinical laboratories systematically and routinely identify, share, and report novel candidate genes. To facilitate this change in practice, we developed criteria for triaging, sharing, and reporting novel candidate genes that are most likely to be promptly validated as underlying a Mendelian condition and translated to use in clinical settings.
Collapse
Affiliation(s)
- Jessica X Chong
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA; Brotman-Baty Institute for Precision Medicine, Seattle, WA.
| | - Seth I Berger
- Center for Genetic Medicine Research, Children's National Research Institute, Washington, DC
| | - Samantha Baxter
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Erica Smith
- Department of Clinical Diagnostics, Ambry Genetics, Aliso Viejo, CA
| | - Changrui Xiao
- Department of Neurology, University of California Irvine, Orange, CA
| | - Daniel G Calame
- Department of Pediatrics, Division of Pediatric Neurology and Developmental Neurosciences, Baylor College of Medicine, Houston, TX
| | | | | | - Stephanie DiTroia
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Michael J Bamshad
- Department of Pediatrics, Division of Genetic Medicine, University of Washington, Seattle, WA; Brotman-Baty Institute for Precision Medicine, Seattle, WA; Department of Pediatrics, Division of Genetic Medicine, Seattle Children's Hospital, Seattle, WA
| | - Heidi L Rehm
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
30
|
Enjeti AK, Walker N, Fahey O, Johnston E, Legge-Wilkinson H, Ramsurrun N, Sillar J, Lincz LF, Ziolkowski A, Mossman D. Certainty in uncertainty: Determining the rate and reasons for reclassification of variants of uncertain significance in haematological malignancies. EJHAEM 2024; 5:957-963. [PMID: 39415915 PMCID: PMC11474286 DOI: 10.1002/jha2.1002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 04/13/2024] [Revised: 08/09/2024] [Accepted: 08/13/2024] [Indexed: 10/19/2024]
Abstract
Introduction Variants of uncertain significance (VUS) are commonly reported in cancer with the widespread adoption of diagnostic massive parallel sequencing. The rate of reclassification of VUS in patients with haematological malignancy is not known and we evaluated this retrospectively. We also investigated whether re-evaluating VUS in 12-24 months or greater than 24 months post-initial classification was significant. Method A retrospective audit of patients with haematological malignancies referred to the Molecular Medicine Department at the John Hunter Hospital in Newcastle, Australia between September 2018 and December 2021. Data was analysed for VUS, which was then re-analysed in standard software using current somatic variant guidelines. Proportions of VUS at baseline were compared to post-re-analysis. Results The most common diagnoses in the patient cohort (n = 944) were acute myelogenous leukaemia (41%), myelodysplastic syndrome (31%), and chronic myelomonocytic leukaemia (7%). A total of 210 VUS were re-analysed. The most common VUS were in the TET2 (20%), RUNX1 (10%) and DNMT3A (9%) genes. A total of 103 were re-analysed at 24-39 months post-initial classification and 107 variants were re-analysed between 12 and 24 months post-initial classification. Of these, 33 (16%) of VUS were re-classified at 24-39 months and 12 (11%) were re-classified at 12-24 months post-initial classification. The most common variants that were re-classified in both groups were CSF3R (32%), TET2 (29%), ASXL1 (11%) and ZRSR2 (11%). Conclusion This study on reclassification of VUS in blood cancers demonstrated that one in seven VUS were re-classified 12 months post initial classification. This can inform practice guidelines and potentially impact the prognosis, diagnosis and treatment of haematological malignancies.
Collapse
Affiliation(s)
- Anoop K Enjeti
- Department of Molecular Medicine NSW Health Pathology, John Hunter Hospital Waratah Australia
- Department of Haematology Calvary Mater Newcastle Waratah Australia
- Precision Medicine Research Program, University of Newcastle Waratah Australia
- School of Medicine and Public Health University of Newcastle Waratah Australia
- Hunter Medical Research Institute Waratah Australia
| | - Natasha Walker
- Precision Medicine Research Program, University of Newcastle Waratah Australia
| | - Oliver Fahey
- Precision Medicine Research Program, University of Newcastle Waratah Australia
| | - Elizabeth Johnston
- Precision Medicine Research Program, University of Newcastle Waratah Australia
| | | | - Nateika Ramsurrun
- Precision Medicine Research Program, University of Newcastle Waratah Australia
| | - Jonathan Sillar
- Department of Haematology Calvary Mater Newcastle Waratah Australia
- Precision Medicine Research Program, University of Newcastle Waratah Australia
- School of Medicine and Public Health University of Newcastle Waratah Australia
- Hunter Medical Research Institute Waratah Australia
| | - Lisa F Lincz
- Department of Haematology Calvary Mater Newcastle Waratah Australia
- School of Medicine and Public Health University of Newcastle Waratah Australia
| | - Andrew Ziolkowski
- Department of Molecular Medicine NSW Health Pathology, John Hunter Hospital Waratah Australia
| | - David Mossman
- Department of Molecular Medicine NSW Health Pathology, John Hunter Hospital Waratah Australia
| |
Collapse
|
31
|
Clarke B, Holtkamp E, Öztürk H, Mück M, Wahlberg M, Meyer K, Munzlinger F, Brechtmann F, Hölzlwimmer FR, Lindner J, Chen Z, Gagneur J, Stegle O. Integration of variant annotations using deep set networks boosts rare variant association testing. Nat Genet 2024; 56:2271-2280. [PMID: 39322779 PMCID: PMC11525182 DOI: 10.1038/s41588-024-01919-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/10/2023] [Accepted: 08/20/2024] [Indexed: 09/27/2024]
Abstract
Rare genetic variants can have strong effects on phenotypes, yet accounting for rare variants in genetic analyses is statistically challenging due to the limited number of allele carriers and the burden of multiple testing. While rich variant annotations promise to enable well-powered rare variant association tests, methods integrating variant annotations in a data-driven manner are lacking. Here we propose deep rare variant association testing (DeepRVAT), a model based on set neural networks that learns a trait-agnostic gene impairment score from rare variant annotations and phenotypes, enabling both gene discovery and trait prediction. On 34 quantitative and 63 binary traits, using whole-exome-sequencing data from UK Biobank, we find that DeepRVAT yields substantial gains in gene discoveries and improved detection of individuals at high genetic risk. Finally, we demonstrate how DeepRVAT enables calibrated and computationally efficient rare variant tests at biobank scale, aiding the discovery of genetic risk factors for human disease traits.
Collapse
Affiliation(s)
- Brian Clarke
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- AI Health Innovation Cluster, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Eva Holtkamp
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Helmholtz Association-Munich School for Data Science (MUDS), Munich, Germany
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany
| | - Hakime Öztürk
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marcel Mück
- AI Health Innovation Cluster, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Magnus Wahlberg
- AI Health Innovation Cluster, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kayla Meyer
- AI Health Innovation Cluster, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Munzlinger
- AI Health Innovation Cluster, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Brechtmann
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- Munich Center for Machine Learning, Munich, Germany
| | - Florian R Hölzlwimmer
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Jonas Lindner
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Zhifen Chen
- Department of Cardiology, Deutsches Herzzentrum München, Technical University Munich, Munich, Germany
- Deutsches Zentrum für Herz- und Kreislaufforschung (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Julien Gagneur
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany.
- Computational Health Center, Helmholtz Center Munich, Neuherberg, Germany.
- Munich Center for Machine Learning, Munich, Germany.
- Institute of Human Genetics, School of Medicine and Health, Technical University of Munich, Munich, Germany.
| | - Oliver Stegle
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, UK.
- Wellcome Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK.
| |
Collapse
|
32
|
Cheng T, Liu Z, Li H, Huang X, Wang W, Shi C, Zhang X, Chen H, Yao Z, Zhao P, Peng X, Sun MX. Sperm-origin paternal effects on root stem cell niche differentiation. Nature 2024; 634:220-227. [PMID: 39198649 DOI: 10.1038/s41586-024-07885-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/07/2023] [Accepted: 07/30/2024] [Indexed: 09/01/2024]
Abstract
Fertilization introduces parental genetic information into the zygote to guide embryogenesis. Parental contributions to postfertilization development have been discussed for decades, and the data available show that both parents contribute to the zygotic transcriptome, suggesting a paternal role in early embryogenesis1-6. However, because the specific paternal effects on postfertilization development and the molecular pathways underpinning these effects remain poorly understood, paternal contribution to early embryogenesis and plant development has not yet been adequately demonstrated7. Here our research shows that TREE1 and its homologue DAZ3 are expressed exclusively in Arabidopsis sperm. Despite presenting no evident defects in sperm development and fertilization, tree1 daz3 unexpectedly led to aberrant differentiation of the embryo root stem cell niche. This defect persisted in seedlings and disrupted root tip regeneration, comparable to congenital defects in animals. TREE1 and DAZ3 function by suppression of maternal RKD2 transcription, thus mitigating the detrimental maternal effects from RKD2 on root stem cell niche. Therefore, our findings illuminate how genetic deficiencies in sperm can exert enduring paternal effects on specific plant organ differentiation and how parental-of-origin genes interact to ensure normal embryogenesis. This work also provides a new concept of how gamete quality or genetic deficiency can affect specific plant organ formation.
Collapse
Affiliation(s)
- Tianhe Cheng
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhenzhen Liu
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Haiming Li
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xiaorong Huang
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Wei Wang
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ce Shi
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xuecheng Zhang
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Hong Chen
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhuang Yao
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Peng Zhao
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Xiongbo Peng
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China
| | - Meng-Xiang Sun
- State Key Laboratory of Hybrid Rice, College of Life Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
33
|
Capalbo A, de Wert G, Mertes H, Klausner L, Coonen E, Spinella F, Van de Velde H, Viville S, Sermon K, Vermeulen N, Lencz T, Carmi S. Screening embryos for polygenic disease risk: a review of epidemiological, clinical, and ethical considerations. Hum Reprod Update 2024; 30:529-557. [PMID: 38805697 PMCID: PMC11369226 DOI: 10.1093/humupd/dmae012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/10/2024] [Revised: 03/25/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND The genetic composition of embryos generated by in vitro fertilization (IVF) can be examined with preimplantation genetic testing (PGT). Until recently, PGT was limited to detecting single-gene, high-risk pathogenic variants, large structural variants, and aneuploidy. Recent advances have made genome-wide genotyping of IVF embryos feasible and affordable, raising the possibility of screening embryos for their risk of polygenic diseases such as breast cancer, hypertension, diabetes, or schizophrenia. Despite a heated debate around this new technology, called polygenic embryo screening (PES; also PGT-P), it is already available to IVF patients in some countries. Several articles have studied epidemiological, clinical, and ethical perspectives on PES; however, a comprehensive, principled review of this emerging field is missing. OBJECTIVE AND RATIONALE This review has four main goals. First, given the interdisciplinary nature of PES studies, we aim to provide a self-contained educational background about PES to reproductive specialists interested in the subject. Second, we provide a comprehensive and critical review of arguments for and against the introduction of PES, crystallizing and prioritizing the key issues. We also cover the attitudes of IVF patients, clinicians, and the public towards PES. Third, we distinguish between possible future groups of PES patients, highlighting the benefits and harms pertaining to each group. Finally, our review, which is supported by ESHRE, is intended to aid healthcare professionals and policymakers in decision-making regarding whether to introduce PES in the clinic, and if so, how, and to whom. SEARCH METHODS We searched for PubMed-indexed articles published between 1/1/2003 and 1/3/2024 using the terms 'polygenic embryo screening', 'polygenic preimplantation', and 'PGT-P'. We limited the review to primary research papers in English whose main focus was PES for medical conditions. We also included papers that did not appear in the search but were deemed relevant. OUTCOMES The main theoretical benefit of PES is a reduction in lifetime polygenic disease risk for children born after screening. The magnitude of the risk reduction has been predicted based on statistical modelling, simulations, and sibling pair analyses. Results based on all methods suggest that under the best-case scenario, large relative risk reductions are possible for one or more diseases. However, as these models abstract several practical limitations, the realized benefits may be smaller, particularly due to a limited number of embryos and unclear future accuracy of the risk estimates. PES may negatively impact patients and their future children, as well as society. The main personal harms are an unindicated IVF treatment, a possible reduction in IVF success rates, and patient confusion, incomplete counselling, and choice overload. The main possible societal harms include discarded embryos, an increasing demand for 'designer babies', overemphasis of the genetic determinants of disease, unequal access, and lower utility in people of non-European ancestries. Benefits and harms will vary across the main potential patient groups, comprising patients already requiring IVF, fertile people with a history of a severe polygenic disease, and fertile healthy people. In the United States, the attitudes of IVF patients and the public towards PES seem positive, while healthcare professionals are cautious, sceptical about clinical utility, and concerned about patient counselling. WIDER IMPLICATIONS The theoretical potential of PES to reduce risk across multiple polygenic diseases requires further research into its benefits and harms. Given the large number of practical limitations and possible harms, particularly unnecessary IVF treatments and discarded viable embryos, PES should be offered only within a research context before further clarity is achieved regarding its balance of benefits and harms. The gap in attitudes between healthcare professionals and the public needs to be narrowed by expanding public and patient education and providing resources for informative and unbiased genetic counselling.
Collapse
Affiliation(s)
- Antonio Capalbo
- Juno Genetics, Department of Reproductive Genetics, Rome, Italy
- Center for Advanced Studies and Technology (CAST), Department of Medical Genetics, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Guido de Wert
- Department of Health, Ethics & Society, CAPHRI-School for Public Health and Primary Care and GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Heidi Mertes
- Department of Philosophy and Moral Sciences, Ghent University, Ghent, Belgium
- Department of Public Health and Primary Care, Ghent University, Ghent, Belgium
| | - Liraz Klausner
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Edith Coonen
- Departments of Clinical Genetics and Reproductive Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands
- School for Oncology and Developmental Biology, GROW, Maastricht University, Maastricht, The Netherlands
| | - Francesca Spinella
- Eurofins GENOMA Group Srl, Molecular Genetics Laboratories, Department of Scientific Communication, Rome, Italy
| | - Hilde Van de Velde
- Research Group Genetics Reproduction and Development (GRAD), Vrije Universiteit Brussel, Brussel, Belgium
- Brussels IVF, UZ Brussel, Brussel, Belgium
| | - Stephane Viville
- Laboratoire de Génétique Médicale LGM, Institut de Génétique Médicale d’Alsace IGMA, INSERM UMR 1112, Université de Strasbourg, France
- Laboratoire de Diagnostic Génétique, Unité de Génétique de l’infertilité (UF3472), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Karen Sermon
- Research Group Genetics Reproduction and Development (GRAD), Vrije Universiteit Brussel, Brussel, Belgium
| | | | - Todd Lencz
- Institute of Behavioral Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA
- Departments of Psychiatry and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Shai Carmi
- Braun School of Public Health and Community Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
34
|
Ye Q, Liu FY, Xia XJ, Chen XY, Zou L, Wu HM, Li DD, Xia CN, Huang T, Cui Y, Zou Y. Whole exome sequencing identifies a novel mutation in Annexin A4 that is associated with recurrent spontaneous abortion. Front Med (Lausanne) 2024; 11:1462649. [PMID: 39399103 PMCID: PMC11466819 DOI: 10.3389/fmed.2024.1462649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/10/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024] Open
Abstract
Background Recurrent spontaneous abortion (RSA) is a multifactorial disease, the exact causes of which are still unknown. Environmental, maternal, and genetic factors have been shown to contribute to this condition. The aim of this study was to investigate the presence of mutations in the ANXA4 gene in patients with RSA. Methods Genomic DNA was extracted from 325 patients with RSA and 941 control women with a normal reproductive history for whole-exome sequencing (WES). The detected variants were annotated and filtered, and the pathogenicity of the variants was predicted through the SIFT online tool, functional enrichment analyses, Sanger sequencing validation, prediction of changes in protein structure, and evolutionary conservation analysis. Furthermore, plasmid construction, Western blotting, RT-qPCR, and cell migration, invasion and adhesion assays were used to detect the effects of ANXA4 mutations on protein function. Results An ANXA4 mutation (p.G8D) in 1 of the 325 samples from patients with RSA (RSA-219) was identified through WES. This mutation was not detected in 941 controls or included in public databases. Evolutionary conservation analysis revealed that the amino acid residue affected by the mutation (p.G8D) was highly conserved among 13 vertebrate species, and the SIFT program and structural modeling analysis predicted that this mutation was harmful. Furthermore, functional assays revealed that this mutation could inhibit cell migration, invasion and adhesion. Conclusion Our study suggests that an unreported novel ANXA4 mutation (p.G8D) plays an important role in the pathogenesis of RSA and may contribute to the genetic diagnosis of RSA.
Collapse
Affiliation(s)
- Qian Ye
- Department of Traditional Chinese Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Research Unit of Female Reproduction with Integrated Chinese and Western Medicine of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Fa-Ying Liu
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Research Unit of Female Reproduction with Integrated Chinese and Western Medicine of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Xiao-Jian Xia
- Department of Traditional Chinese Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Research Unit of Female Reproduction with Integrated Chinese and Western Medicine of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Xiao-Yong Chen
- Department of Traditional Chinese Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Research Unit of Female Reproduction with Integrated Chinese and Western Medicine of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Li Zou
- Quality Control Office, Ganzhou People's Hospital, Ganzhou, China
| | - Hui-Min Wu
- Graduate School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Dan-Dan Li
- Graduate School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Chen-Nian Xia
- Graduate School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Ting Huang
- Graduate School of Clinical Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Ying Cui
- Department of Traditional Chinese Medicine, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Research Unit of Female Reproduction with Integrated Chinese and Western Medicine of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Yang Zou
- Key Laboratory of Women's Reproductive Health of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Key Research Unit of Female Reproduction with Integrated Chinese and Western Medicine of Jiangxi Province, Jiangxi Maternal and Child Health Hospital, Nanchang, China
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| |
Collapse
|
35
|
Luan M, Chen K, Zhao W, Tang M, Wang L, Liu S, Zhu L, Xie S. Selective Effect of DNA N6-Methyladenosine Modification on Transcriptional Genetic Variations in East Asian Samples. Int J Mol Sci 2024; 25:10400. [PMID: 39408729 PMCID: PMC11477068 DOI: 10.3390/ijms251910400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/23/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Genetic variations and DNA modification are two common dominant factors ubiquitous across the entire human genome and induce human disease, especially through static genetic variations in DNA or RNA that cause human genetic diseases. DNA N6-methyladenosine (6mA) methylation, as a new epigenetic modification mark, has been widely studied for regulatory biological processes in humans. However, the effect of DNA modification on dynamic transcriptional genetic variations from DNA to RNA has rarely been reported. Here, we identified DNA, RNA and transcriptional genetic variations from Illumina short-read sequencing data in East Asian samples (HX1 and AK1) and detected global DNA 6mA modification using single-molecule, real-time sequencing (SMRT) data. We decoded the effects of DNA 6mA modification on transcriptional genetic variations in East Asian samples and the results were extensively verified in the HeLa cell line. DNA 6mA modification had a stabilized distribution in the East Asian samples and the methylated genes were less likely to mutate than the non-methylated genes. For methylated genes, the 6mA density was positively correlated with the number of variations. DNA 6mA modification had a selective effect on transcriptional genetic variations from DNA to RNA, in which the dynamic transcriptional variations of heterozygous (0/1 to 0/1) and homozygous (1/1 to 1/1) were significantly affected by 6mA modification. The effect of DNA methylation on transcriptional genetic variations provides new insights into the influencing factors of DNA to RNA transcriptional regulation in the central doctrine of molecular biology.
Collapse
Affiliation(s)
- Meiwei Luan
- School of Basic Medicine, Harbin Medical University, Harbin 150081, China;
| | - Kaining Chen
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 511436, China;
| | - Wenwen Zhao
- College of Forestry, Hainan University, Haikou 570228, China; (W.Z.); (M.T.); (L.W.); (S.L.)
| | - Minqiang Tang
- College of Forestry, Hainan University, Haikou 570228, China; (W.Z.); (M.T.); (L.W.); (S.L.)
| | - Lingxia Wang
- College of Forestry, Hainan University, Haikou 570228, China; (W.Z.); (M.T.); (L.W.); (S.L.)
| | - Shoubai Liu
- College of Forestry, Hainan University, Haikou 570228, China; (W.Z.); (M.T.); (L.W.); (S.L.)
| | - Linan Zhu
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99163, USA;
| | - Shangqian Xie
- College of Forestry, Hainan University, Haikou 570228, China; (W.Z.); (M.T.); (L.W.); (S.L.)
| |
Collapse
|
36
|
Griffith EC, West AE, Greenberg ME. Neuronal enhancers fine-tune adaptive circuit plasticity. Neuron 2024; 112:3043-3057. [PMID: 39208805 PMCID: PMC11550865 DOI: 10.1016/j.neuron.2024.08.002] [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] [Academic Contribution Register] [Received: 05/01/2023] [Revised: 07/22/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Neuronal activity-regulated gene expression plays a crucial role in sculpting neural circuits that underpin adaptive brain function. Transcriptional enhancers are now recognized as key components of gene regulation that orchestrate spatiotemporally precise patterns of gene transcription. We propose that the dynamics of enhancer activation uniquely position these genomic elements to finely tune activity-dependent cellular plasticity. Enhancer specificity and modularity can be exploited to gain selective genetic access to specific cell states, and the precise modulation of target gene expression within restricted cellular contexts enabled by targeted enhancer manipulation allows for fine-grained evaluation of gene function. Mounting evidence also suggests that enduring stimulus-induced changes in enhancer states can modify target gene activation upon restimulation, thereby contributing to a form of cell-wide metaplasticity. We advocate for focused exploration of activity-dependent enhancer function to gain new insight into the mechanisms underlying brain plasticity and cognitive dysfunction.
Collapse
Affiliation(s)
- Eric C Griffith
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| | | |
Collapse
|
37
|
Wen W, Zhong J, Zhang Z, Jia L, Chu T, Wang N, Danko CG, Wang Z. dHICA: a deep transformer-based model enables accurate histone imputation from chromatin accessibility. Brief Bioinform 2024; 25:bbae459. [PMID: 39316943 PMCID: PMC11421843 DOI: 10.1093/bib/bbae459] [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] [Academic Contribution Register] [Received: 05/16/2024] [Revised: 07/13/2024] [Accepted: 09/04/2024] [Indexed: 09/26/2024] Open
Abstract
Histone modifications (HMs) are pivotal in various biological processes, including transcription, replication, and DNA repair, significantly impacting chromatin structure. These modifications underpin the molecular mechanisms of cell-type-specific gene expression and complex diseases. However, annotating HMs across different cell types solely using experimental approaches is impractical due to cost and time constraints. Herein, we present dHICA (deep histone imputation using chromatin accessibility), a novel deep learning framework that integrates DNA sequences and chromatin accessibility data to predict multiple HM tracks. Employing the transformer architecture alongside dilated convolutions, dHICA boasts an extensive receptive field and captures more cell-type-specific information. dHICA outperforms state-of-the-art baselines and achieves superior performance in cell-type-specific loci and gene elements, aligning with biological expectations. Furthermore, dHICA's imputations hold significant potential for downstream applications, including chromatin state segmentation and elucidating the functional implications of SNPs (Single Nucleotide Polymorphisms). In conclusion, dHICA serves as a valuable tool for advancing the understanding of chromatin dynamics, offering enhanced predictive capabilities and interpretability.
Collapse
Affiliation(s)
- Wen Wen
- School of Software Technology, Dalian University of Technology, Linggong Rd, Liaoning 116024, China
| | - Jiaxin Zhong
- School of Software Technology, Dalian University of Technology, Linggong Rd, Liaoning 116024, China
| | - Zhaoxi Zhang
- School of Software Technology, Dalian University of Technology, Linggong Rd, Liaoning 116024, China
| | - Lijuan Jia
- School of Software Technology, Dalian University of Technology, Linggong Rd, Liaoning 116024, China
| | - Tinyi Chu
- Meinig School of Biomedical Engineering, Cornell University, Weill Hall, Ithaca, NY 14853, United States
| | - Nating Wang
- Department of Molecular Biology and Genetics, Cornell University, Biotechnology Building, Ithaca, NY 14853, United States
| | - Charles G Danko
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Hungerford Hill Rd, Ithaca, NY 14853, United States
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Tower Rd, Ithaca, NY 14853, United States
| | - Zhong Wang
- School of Software Technology, Dalian University of Technology, Linggong Rd, Liaoning 116024, China
| |
Collapse
|
38
|
Yalcouyé A, Dabitao D, Samassékou O, Nembaware V, Kané F, Alimohamed MZ, El-Kamah G, Mutesa L, Ndiaye R, Ramsay M, Doumbia S, Williams S, Traoré M, Wonkam A, Landouré G. Human genetics and genomics as a unifying factor for harmony and progress in Africa: a report from the 12 th African Society of Human Genetics meeting in Bamako, Mali. Pan Afr Med J 2024; 49:19. [PMID: 39711838 PMCID: PMC11662216 DOI: 10.11604/pamj.2024.49.19.41189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/24/2023] [Accepted: 07/30/2023] [Indexed: 12/24/2024] Open
Abstract
Since its inception in 2003, the African Society of Human Genetics (AfSHG) has been central to the promotion of genetics research on the continent, and facilitated the networking of African researchers within Africa and abroad, thereby significantly contributing to the career development of African geneticists. The continuation of these accomplishments was stimulated by the 12th international conference of AfSHG held jointly with the 1st Congress of the Malian Society of Human Genetics (MSHG) in Bamako, Mali from September 18th to 21st 2019. The main theme of the conference was "Human Genetics and Genomics as a Unifying Factor for Harmony and Progress in Africa". The goals of the meeting were to promote the work conducted mainly by African researchers and to contribute to scientific knowledge through genetic research. Despite challenges due to security issues in Mali, this conference attracted many scientists, including key experts in genetics and associated fields, making the conference successful scientifically and geographically. Overall, 172 delegates from 24 countries attended. Sessions on various topics relevant to Africa were held. These included the genetics of infectious diseases, cancer, and rare diseases as well as bioinformatics, pharmacogenomics, population genetics, and ethical, legal, and social issues, particularly with respect to genetic research in African populations. The need for genetic data sharing to improve research and health and the focus of actionable research for African populations was stressed throughout the meeting.
Collapse
Affiliation(s)
- Abdoulaye Yalcouyé
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Djénéba Dabitao
- Faculté de Pharmacie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Oumar Samassékou
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Victoria Nembaware
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Fousséyni Kané
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Mohamed Zahir Alimohamed
- Department of Haematology and Blood Transfusion, Muhimbili University of Health and Allied Sciences, Dar-es-Salaam, Tanzania
| | - Ghada El-Kamah
- Human Genetics and Genome Research Division, Centre of Scientific Excellence of Human Genetics, National Research Centre, Cairo, Egypt
| | - Leon Mutesa
- Centre for Human Genetics, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Rokhaya Ndiaye
- Faculty of Medicine, Pharmacy and Dentistry, Cheikh Anta Diop University, Dakar, Senegal
| | - Michele Ramsay
- Division of Human Genetics, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Seydou Doumbia
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Scott Williams
- Departments of Population and Quantitative Health Sciences, and Genetics and Genome Sciences, Cleveland Institute of Computational Biology, Case Western Reserve University, Cleveland, Ohi
| | - Mahamadou Traoré
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
| | - Ambroise Wonkam
- Division of Human Genetics, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- McKusick-Nathans Institute of Genetic Medicine and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guida Landouré
- Faculté de Médecine et d’Odontostomatologie, Université des Sciences, des Techniques et des Technologies de Bamako, Bamako, Mali
- Service de Neurologie, Centre Hospitalier Universitaire du Point G, Bamako, Mali
| |
Collapse
|
39
|
Gracheva AS, Kashatnikova DA, Redkin IV, Zakharchenko VE, Kuzovlev AN, Salnikova LE. Genetics and Traumatic Brain Injury: Findings from an Exome-Based Study of a 50-Patient Case Series. Curr Issues Mol Biol 2024; 46:10351-10368. [PMID: 39329968 PMCID: PMC11430351 DOI: 10.3390/cimb46090616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/21/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
Traumatic brain injury (TBI) is the leading cause of global mortality and morbidity. Because TBI is accident-related, the role of genetics in predisposing to TBI has been largely unexplored. However, the likelihood of injury may not be entirely random and may be associated with certain physical and mental characteristics. In this study, we analyzed the exomes of 50 patients undergoing rehabilitation after TBI. Patients were divided into three groups according to rehabilitation outcome: improvement, no change, and deterioration/death. We focused on rare, potentially functional missense and high-impact variants in genes intolerant to these variants. The concordant results from the three independent groups of patients allowed for the suggestion of the existence of a genetic predisposition to TBI, associated with rare functional variations in intolerant genes, with a prevalent dominant mode of inheritance and neurological manifestations in the genetic phenotypes according to the OMIM database. Forty-four of the 50 patients had one or more rare, potentially deleterious variants in one or more neurological genes. Comparison of these results with those of a 50-sampled matched non-TBI cohort revealed significant differences: P = 2.6 × 10-3, OR = 4.89 (1.77-13.47). There were no differences in the distribution of the genes of interest between the TBI patient groups. Our exploratory study provides new insights into the impact of genetics on TBI risk and is the first to address potential genetic susceptibility to TBI.
Collapse
Affiliation(s)
- Alesya S Gracheva
- The Department of Population Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Darya A Kashatnikova
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- The Laboratory of Molecular Pathophysiology, Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Ivan V Redkin
- The Laboratory of Organoprotection in Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Vladislav E Zakharchenko
- The Department of Clinical Laboratory Diagnostics, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Artem N Kuzovlev
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
| | - Lyubov E Salnikova
- The Laboratory of Clinical Pathophysiology of Critical Conditions, Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 107031 Moscow, Russia
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, 119991 Moscow, Russia
- The Laboratory of Molecular Immunology, National Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia
| |
Collapse
|
40
|
Madeo AC, Kohlmann W, Liao Y, Zhong L, Rothwell E, Kaphingst KA. Women's preferences for genetic screening in routine care: A qualitative study. PATIENT EDUCATION AND COUNSELING 2024; 130:108439. [PMID: 39303503 DOI: 10.1016/j.pec.2024.108439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 02/26/2024] [Revised: 08/06/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE Examine decision-making regarding when women would prefer to receive reproductive carrier and cancer predisposition screening and from what clinician. METHODS 20 women completed in-depth interviews via Zoom exploring their views on the provision of reproductive carrier and cancer predisposition screening. Our analysis identified themes related to what informs women's preferences for when they would like to receive a genetic screening offer and by which clinician. RESULTS Participants' responses to questions about when they would be interested in receiving genetic screening were best understood through the lens of the Extended Parallel Process Model. Specifically, personal utility of the information, a woman's family health history and cost were key factors in decision-making. Women considered their clinician's knowledge and their trust in and relationship with the clinician when deciding from whom they would prefer to receive an offer of genetic screening. CONCLUSION OB/GYN clinic patients may accept an offer of genetic screening from a knowledgeable and trusted clinician for carrier and cancer predisposition screening preconceptionally or prenatally. PRACTICE IMPLICATIONS Integrating genetic reproductive and cancer predisposition screening into the care provided to reproductive age OB/GYN patients may be acceptable to this population.
Collapse
Affiliation(s)
- Anne C Madeo
- Huntsman Cancer Institute, University of Utah Health, Salt Lake City, UT, USA; Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA.
| | - Wendy Kohlmann
- Huntsman Cancer Institute, University of Utah Health, Salt Lake City, UT, USA
| | - Yi Liao
- Huntsman Cancer Institute, University of Utah Health, Salt Lake City, UT, USA; Department of Communication, University of Utah, Salt Lake City, UT, USA
| | - Lingzi Zhong
- Department of Communication, University of Utah, Salt Lake City, UT, USA
| | - Erin Rothwell
- Department of Obstetrics and Gynecology, University of Utah, Salt Lake City, UT, USA
| | - Kimberly A Kaphingst
- Huntsman Cancer Institute, University of Utah Health, Salt Lake City, UT, USA; Department of Communication, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
41
|
Sundaram L, Kumar A, Zatzman M, Salcedo A, Ravindra N, Shams S, Louie BH, Bagdatli ST, Myers MA, Sarmashghi S, Choi HY, Choi WY, Yost KE, Zhao Y, Granja JM, Hinoue T, Hayes DN, Cherniack A, Felau I, Choudhry H, Zenklusen JC, Farh KKH, McPherson A, Curtis C, Laird PW, Demchok JA, Yang L, Tarnuzzer R, Caesar-Johnson SJ, Wang Z, Doane AS, Khurana E, Castro MAA, Lazar AJ, Broom BM, Weinstein JN, Akbani R, Kumar SV, Raphael BJ, Wong CK, Stuart JM, Safavi R, Benz CC, Johnson BK, Kyi C, Shen H, Corces MR, Chang HY, Greenleaf WJ. Single-cell chromatin accessibility reveals malignant regulatory programs in primary human cancers. Science 2024; 385:eadk9217. [PMID: 39236169 DOI: 10.1126/science.adk9217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/19/2023] [Accepted: 07/03/2024] [Indexed: 09/07/2024]
Abstract
To identify cancer-associated gene regulatory changes, we generated single-cell chromatin accessibility landscapes across eight tumor types as part of The Cancer Genome Atlas. Tumor chromatin accessibility is strongly influenced by copy number alterations that can be used to identify subclones, yet underlying cis-regulatory landscapes retain cancer type-specific features. Using organ-matched healthy tissues, we identified the "nearest healthy" cell types in diverse cancers, demonstrating that the chromatin signature of basal-like-subtype breast cancer is most similar to secretory-type luminal epithelial cells. Neural network models trained to learn regulatory programs in cancer revealed enrichment of model-prioritized somatic noncoding mutations near cancer-associated genes, suggesting that dispersed, nonrecurrent, noncoding mutations in cancer are functional. Overall, these data and interpretable gene regulatory models for cancer and healthy tissue provide a framework for understanding cancer-specific gene regulation.
Collapse
Affiliation(s)
- Laksshman Sundaram
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Illumina AI laboratory, Illumina Inc, Foster City, CA, USA
- NVIDIA Bio Research, NVIDIA, Santa Clara, CA, USA
| | - Arvind Kumar
- Illumina AI laboratory, Illumina Inc, Foster City, CA, USA
| | - Matthew Zatzman
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Neal Ravindra
- Illumina AI laboratory, Illumina Inc, Foster City, CA, USA
| | - Shadi Shams
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Bryan H Louie
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - S Tansu Bagdatli
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Matthew A Myers
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Hyo Young Choi
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Won-Young Choi
- UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kathryn E Yost
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Yanding Zhao
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Jeffrey M Granja
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Toshinori Hinoue
- Center for Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - D Neil Hayes
- Department of Preventive Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- UTHSC Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | - Ina Felau
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jean C Zenklusen
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Andrew McPherson
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christina Curtis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Peter W Laird
- Center for Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - John A Demchok
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Liming Yang
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Roy Tarnuzzer
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | | | - Zhining Wang
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, NIH, 9609 Medical Center Drive, Rockville, MD 20850, USA
| | - Ashley S Doane
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ekta Khurana
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10065, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Mauro A A Castro
- Bioinformatics and Systems Biology Laboratory, Federal University of Paraná, Curitiba 81520-260, Brazil
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bradley M Broom
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - John N Weinstein
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Rehan Akbani
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shwetha V Kumar
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, 35 Olden Street, Princeton, NJ 08540
| | - Christopher K Wong
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Joshua M Stuart
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Rojin Safavi
- Biomolecular Engineering Department, School of Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | | | - Benjamin K Johnson
- Center for Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Cindy Kyi
- Center for Cancer Genomics, National Cancer Institute, Bethesda, MD 20892, USA
| | - Hui Shen
- Center for Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - M Ryan Corces
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Gladstone Institute of Neurological Disease, Gladstone Institutes, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Howard Y Chang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
- Howard Hughes Medical Institute, Stanford University, School of Medicine, Stanford, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| |
Collapse
|
42
|
Thaxton C, Biesecker LG, DiStefano M, Haendel M, Hamosh A, Owens E, Plon SE, Rehm HL, Berg JS. Implementation of a dyadic nomenclature for monogenic diseases. Am J Hum Genet 2024; 111:1810-1818. [PMID: 39241757 PMCID: PMC11393707 DOI: 10.1016/j.ajhg.2024.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/31/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 09/09/2024] Open
Abstract
A core task when establishing the strength of evidence for a gene's role in a monogenic disorder is determining the appropriate disease entity to curate. Establishing this concept determines which evidence can be applied and quantified toward the final gene-disease validity, variant pathogenicity, or actionability classification. Genes with implications in more than one phenotype can necessitate a process of lumping and splitting, disease reorganization, and updates to disease nomenclature. Reappraisal of the names that are used as labels for disease entities is therefore a necessary and perpetual process. The Clinical Genome Resource (ClinGen), in collaboration with representatives from Monarch Disease Ontology (Mondo) and Online Inheritance in Man (OMIM), formed the Disease Naming Advisory Committee (DNAC) to develop guidance for groups faced with the need to establish the "curated disease entity" for gene-phenotype validity and variant pathogenicity and to update disease names for clinical use when necessary. The objective of this group was to harmonize guidance for disease naming across these nosologic entities and among ClinGen curation groups in collaboration with other disease-related professional groups. Here, we present the initial guidance developed by the DNAC with representative examples provided by the ClinGen expert panels and working groups that warranted nomenclature updates. We also discuss the broader implications of these efforts and their benefits for harmonization of gene-disease validity curation. Overall, this work sheds light on current inconsistencies and/or discrepancies and is designed to engage the broader community on how ClinGen defines monogenic disorders using a consistent approach for disease naming.
Collapse
Affiliation(s)
- Courtney Thaxton
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Leslie G Biesecker
- Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marina DiStefano
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Melissa Haendel
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ada Hamosh
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Emma Owens
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Sharon E Plon
- Department of Pediatrics/Hematology-Oncology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Heidi L Rehm
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jonathan S Berg
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
43
|
Chen JH, Landback P, Arsala D, Guzzetta A, Xia S, Atlas J, Sosa D, Zhang YE, Cheng J, Shen B, Long M. Evolutionarily new genes in humans with disease phenotypes reveal functional enrichment patterns shaped by adaptive innovation and sexual selection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.14.567139. [PMID: 38045239 PMCID: PMC10690195 DOI: 10.1101/2023.11.14.567139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/05/2023]
Abstract
New genes (or young genes) are genetic novelties pivotal in mammalian evolution. However, their phenotypic impacts and evolutionary patterns over time remain elusive in humans due to the technical and ethical complexities of functional studies. Integrating gene age dating with Mendelian disease phenotyping, our research shows a gradual rise in disease gene proportion as gene age increases. Logistic regression modeling indicates that this increase in older genes may be related to their longer sequence lengths and higher burdens of deleterious de novo germline variants (DNVs). We also find a steady integration of new genes with biomedical phenotypes into the human genome over macroevolutionary timescales (~0.07% per million years). Despite this stable pace, we observe distinct patterns in phenotypic enrichment, pleiotropy, and selective pressures across gene ages. Notably, young genes show significant enrichment in diseases related to the male reproductive system, indicating strong sexual selection. Young genes also exhibit disease-related functions in tissues and systems potentially linked to human phenotypic innovations, such as increased brain size, musculoskeletal phenotypes, and color vision. We further reveal a logistic growth pattern of pleiotropy over evolutionary time, indicating a diminishing marginal growth of new functions for older genes due to intensifying selective constraints over time. We propose a "pleiotropy-barrier" model that delineates higher potentials for phenotypic innovation in young genes compared to older genes, a process that is subject to natural selection. Our study demonstrates that evolutionarily new genes are critical in influencing human reproductive evolution and adaptive phenotypic innovations driven by sexual and natural selection, with low pleiotropy as a selective advantage.
Collapse
Affiliation(s)
- Jian-Hai Chen
- Department of Ecology and Evolution, The University of Chicago, 1101 E 57th Street, Chicago, IL 60637
- Institutes for Systems Genetics, West China University Hospital, Chengdu 610041, China
| | - Patrick Landback
- Department of Ecology and Evolution, The University of Chicago, 1101 E 57th Street, Chicago, IL 60637
| | - Deanna Arsala
- Department of Ecology and Evolution, The University of Chicago, 1101 E 57th Street, Chicago, IL 60637
| | - Alexander Guzzetta
- Department of Pathology, The University of Chicago, 1101 E 57th Street, Chicago, IL 60637
| | - Shengqian Xia
- Department of Ecology and Evolution, The University of Chicago, 1101 E 57th Street, Chicago, IL 60637
| | - Jared Atlas
- Department of Ecology and Evolution, The University of Chicago, 1101 E 57th Street, Chicago, IL 60637
| | - Dylan Sosa
- Department of Ecology and Evolution, The University of Chicago, 1101 E 57th Street, Chicago, IL 60637
| | - Yong E. Zhang
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jingqiu Cheng
- Institutes for Systems Genetics, West China University Hospital, Chengdu 610041, China
| | - Bairong Shen
- Institutes for Systems Genetics, West China University Hospital, Chengdu 610041, China
| | - Manyuan Long
- Department of Ecology and Evolution, The University of Chicago, 1101 E 57th Street, Chicago, IL 60637
| |
Collapse
|
44
|
Poortman Y, Ens-Dokkum M, Nippert I. The Role of Patient Organizations in Shaping Research, Health Policies, and Health Services for Rare Genetic Diseases: The Dutch Experience. Genes (Basel) 2024; 15:1162. [PMID: 39336753 PMCID: PMC11431757 DOI: 10.3390/genes15091162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/18/2024] [Revised: 08/15/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
In 2023, the genetics scientific community celebrated two special anniversaries: the discovery of the double helix structure of DNA was published in 1953 and in 2003 the Human Genome Project was declared completed and made publicly available. To this day, genetics and genomics research is continuing to evolve at high pace and is identifying a steadily increasing number of genes as causal for distinct genetic diseases. The success story of genetics and genomics would not be complete without taking due account of the role of patient advocacy organizations in this process. This paper is based on the personal narrative (oral history) of a father whose daughter was born with a rare genetic disease (RGD) in the 1960s. The first-hand experience of living as a family with an RGD in those days made him a leading pioneer not only in the foundation of patient organizations at national, pan-European, and international levels but also in the development of multi-stakeholder co-operation and networking. Today, patient advocacy organizations play an active role in shaping health and research policies at national, EU, and international levels to ensure that their needs in regard to advancing RGD diagnostics, care, and treatment are addressed.
Collapse
Affiliation(s)
| | - Martina Ens-Dokkum
- Kentalis International Foundation, 2716 NR Zoetermeer, The Netherlands;
- Curium-Leiden University Medical Center, 2342 AK Oegstgeest, The Netherlands
| | - Irmgard Nippert
- Faculty of Medicine, University of Münster, 48149 Münster, Germany
| |
Collapse
|
45
|
Engreitz JM, Lawson HA, Singh H, Starita LM, Hon GC, Carter H, Sahni N, Reddy TE, Lin X, Li Y, Munshi NV, Chahrour MH, Boyle AP, Hitz BC, Mortazavi A, Craven M, Mohlke KL, Pinello L, Wang T, Kundaje A, Yue F, Cody S, Farrell NP, Love MI, Muffley LA, Pazin MJ, Reese F, Van Buren E, Dey KK, Kircher M, Ma J, Radivojac P, Balliu B, Williams BA, Huangfu D, Park CY, Quertermous T, Das J, Calderwood MA, Fowler DM, Vidal M, Ferreira L, Mooney SD, Pejaver V, Zhao J, Gazal S, Koch E, Reilly SK, Sunyaev S, Carpenter AE, Buenrostro JD, Leslie CS, Savage RE, Giric S, Luo C, Plath K, Barrera A, Schubach M, Gschwind AR, Moore JE, Ahituv N, Yi SS, Hallgrimsdottir I, Gaulton KJ, Sakaue S, Booeshaghi S, Mattei E, Nair S, Pachter L, Wang AT, Shendure J, Agarwal V, Blair A, Chalkiadakis T, Chardon FM, Dash PM, Deng C, Hamazaki N, Keukeleire P, Kubo C, Lalanne JB, Maass T, Martin B, McDiarmid TA, Nobuhara M, Page NF, Regalado S, Sims J, Ushiki A, Best SM, Boyle G, Camp N, Casadei S, Da EY, Dawood M, Dawson SC, Fayer S, Hamm A, James RG, Jarvik GP, McEwen AE, Moore N, Pendyala S, Popp NA, Post M, Rubin AF, Smith NT, Stone J, Tejura M, Wang ZR, Wheelock MK, Woo I, Zapp BD, Amgalan D, Aradhana A, Arana SM, Bassik MC, Bauman JR, Bhattacharya A, Cai XS, Chen Z, Conley S, Deshpande S, Doughty BR, Du PP, Galante JA, Gifford C, Greenleaf WJ, Guo K, Gupta R, Isobe S, Jagoda E, Jain N, Jones H, Kang HY, Kim SH, Kim Y, Klemm S, Kundu R, Kundu S, Lago-Docampo M, Lee-Yow YC, Levin-Konigsberg R, Li DY, Lindenhofer D, Ma XR, Marinov GK, Martyn GE, McCreery CV, Metzl-Raz E, Monteiro JP, Montgomery MT, Mualim KS, Munger C, Munson G, Nguyen TC, Nguyen T, Palmisano BT, Pampari A, Rabinovitch M, Ramste M, Ray J, Roy KR, Rubio OM, Schaepe JM, Schnitzler G, Schreiber J, Sharma D, Sheth MU, Shi H, Singh V, Sinha R, Steinmetz LM, Tan J, Tan A, Tycko J, Valbuena RC, Amiri VVP, van Kooten MJFM, Vaughan-Jackson A, Venida A, Weldy CS, Worssam MD, Xia F, Yao D, Zeng T, Zhao Q, Zhou R, Chen ZS, Cimini BA, Coppin G, Coté AG, Haghighi M, Hao T, Hill DE, Lacoste J, Laval F, Reno C, Roth FP, Singh S, Spirohn-Fitzgerald K, Taipale M, Teelucksingh T, Tixhon M, Yadav A, Yang Z, Kraus WL, Armendariz DA, Dederich AE, Gogate A, El Hayek L, Goetsch SC, Kaur K, Kim HB, McCoy MK, Nzima MZ, Pinzón-Arteaga CA, Posner BA, Schmitz DA, Sivakumar S, Sundarrajan A, Wang L, Wang Y, Wu J, Xu L, Xu J, Yu L, Zhang Y, Zhao H, Zhou Q, Won H, Bell JL, Broadaway KA, Degner KN, Etheridge AS, Koller BH, Mah W, Mu W, Ritola KD, Rosen JD, Schoenrock SA, Sharp RA, Bauer D, Lettre G, Sherwood R, Becerra B, Blaine LJ, Che E, Francoeur MJ, Gibbs EN, Kim N, King EM, Kleinstiver BP, Lecluze E, Li Z, Patel ZM, Phan QV, Ryu J, Starr ML, Wu T, Gersbach CA, Crawford GE, Allen AS, Majoros WH, Iglesias N, Rai R, Venukuttan R, Li B, Anglen T, Bounds LR, Hamilton MC, Liu S, McCutcheon SR, McRoberts Amador CD, Reisman SJ, ter Weele MA, Bodle JC, Streff HL, Siklenka K, Strouse K, Bernstein BE, Babu J, Corona GB, Dong K, Duarte FM, Durand NC, Epstein CB, Fan K, Gaskell E, Hall AW, Ham AM, Knudson MK, Shoresh N, Wekhande S, White CM, Xi W, Satpathy AT, Corces MR, Chang SH, Chin IM, Gardner JM, Gardell ZA, Gutierrez JC, Johnson AW, Kampman L, Kasowski M, Lareau CA, Liu V, Ludwig LS, McGinnis CS, Menon S, Qualls A, Sandor K, Turner AW, Ye CJ, Yin Y, Zhang W, Wold BJ, Carilli M, Cheong D, Filibam G, Green K, Kawauchi S, Kim C, Liang H, Loving R, Luebbert L, MacGregor G, Merchan AG, Rebboah E, Rezaie N, Sakr J, Sullivan DK, Swarna N, Trout D, Upchurch S, Weber R, Castro CP, Chou E, Feng F, Guerra A, Huang Y, Jiang L, Liu J, Mills RE, Qian W, Qin T, Sartor MA, Sherpa RN, Wang J, Wang Y, Welch JD, Zhang Z, Zhao N, Mukherjee S, Page CD, Clarke S, Doty RW, Duan Y, Gordan R, Ko KY, Li S, Li B, Thomson A, Raychaudhuri S, Price A, Ali TA, Dey KK, Durvasula A, Kellis M, Iakoucheva LM, Kakati T, Chen Y, Benazouz M, Jain S, Zeiberg D, De Paolis Kaluza MC, Velyunskiy M, Gasch A, Huang K, Jin Y, Lu Q, Miao J, Ohtake M, Scopel E, Steiner RD, Sverchkov Y, Weng Z, Garber M, Fu Y, Haas N, Li X, Phalke N, Shan SC, Shedd N, Yu T, Zhang Y, Zhou H, Battle A, Jerby L, Kotler E, Kundu S, Marderstein AR, Montgomery SB, Nigam A, Padhi EM, Patel A, Pritchard J, Raine I, Ramalingam V, Rodrigues KB, Schreiber JM, Singhal A, Sinha R, Wang AT, Abundis M, Bisht D, Chakraborty T, Fan J, Hall DR, Rarani ZH, Jain AK, Kaundal B, Keshari S, McGrail D, Pease NA, Yi VF, Wu H, Kannan S, Song H, Cai J, Gao Z, Kurzion R, Leu JI, Li F, Liang D, Ming GL, Musunuru K, Qiu Q, Shi J, Su Y, Tishkoff S, Xie N, Yang Q, Yang W, Zhang H, Zhang Z, Beer MA, Hadjantonakis AK, Adeniyi S, Cho H, Cutler R, Glenn RA, Godovich D, Hu N, Jovanic S, Luo R, Oh JW, Razavi-Mohseni M, Shigaki D, Sidoli S, Vierbuchen T, Wang X, Williams B, Yan J, Yang D, Yang Y, Sander M, Gaulton KJ, Ren B, Bartosik W, Indralingam HS, Klie A, Mummey H, Okino ML, Wang G, Zemke NR, Zhang K, Zhu H, Zaitlen N, Ernst J, Langerman J, Li T, Sun Y, Rudensky AY, Periyakoil PK, Gao VR, Smith MH, Thomas NM, Donlin LT, Lakhanpal A, Southard KM, Ardy RC, Cherry JM, Gerstein MB, Andreeva K, Assis PR, Borsari B, Douglass E, Dong S, Gabdank I, Graham K, Jolanki O, Jou J, Kagda MS, Lee JW, Li M, Lin K, Miyasato SR, Rozowsky J, Small C, Spragins E, Tanaka FY, Whaling IM, Youngworth IA, Sloan CA, Belter E, Chen X, Chisholm RL, Dickson P, Fan C, Fulton L, Li D, Lindsay T, Luan Y, Luo Y, Lyu H, Ma X, Macias-Velasco J, Miga KH, Quaid K, Stitziel N, Stranger BE, Tomlinson C, Wang J, Zhang W, Zhang B, Zhao G, Zhuo X, Brennand K, Ciccia A, Hayward SB, Huang JW, Leuzzi G, Taglialatela A, Thakar T, Vaitsiankova A, Dey KK, Ali TA, Kim A, Grimes HL, Salomonis N, Gupta R, Fang S, Lee-Kim V, Heinig M, Losert C, Jones TR, Donnard E, Murphy M, Roberts E, Song S, Mostafavi S, Sasse A, Spiro A, Pennacchio LA, Kato M, Kosicki M, Mannion B, Slaven N, Visel A, Pollard KS, Drusinsky S, Whalen S, Ray J, Harten IA, Ho CH, Sanjana NE, Caragine C, Morris JA, Seruggia D, Kutschat AP, Wittibschlager S, Xu H, Fu R, He W, Zhang L, Osorio D, Bly Z, Calluori S, Gilchrist DA, Hutter CM, Morris SA, Samer EK. Deciphering the impact of genomic variation on function. Nature 2024; 633:47-57. [PMID: 39232149 DOI: 10.1038/s41586-024-07510-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/11/2023] [Accepted: 05/02/2024] [Indexed: 09/06/2024]
Abstract
Our genomes influence nearly every aspect of human biology-from molecular and cellular functions to phenotypes in health and disease. Studying the differences in DNA sequence between individuals (genomic variation) could reveal previously unknown mechanisms of human biology, uncover the basis of genetic predispositions to diseases, and guide the development of new diagnostic tools and therapeutic agents. Yet, understanding how genomic variation alters genome function to influence phenotype has proved challenging. To unlock these insights, we need a systematic and comprehensive catalogue of genome function and the molecular and cellular effects of genomic variants. Towards this goal, the Impact of Genomic Variation on Function (IGVF) Consortium will combine approaches in single-cell mapping, genomic perturbations and predictive modelling to investigate the relationships among genomic variation, genome function and phenotypes. IGVF will create maps across hundreds of cell types and states describing how coding variants alter protein activity, how noncoding variants change the regulation of gene expression, and how such effects connect through gene-regulatory and protein-interaction networks. These experimental data, computational predictions and accompanying standards and pipelines will be integrated into an open resource that will catalyse community efforts to explore how our genomes influence biology and disease across populations.
Collapse
|
46
|
Kwait R, Pinsky ML, Gignoux‐Wolfsohn S, Eskew EA, Kerwin K, Maslo B. Impact of putatively beneficial genomic loci on gene expression in little brown bats ( Myotis lucifugus, Le Conte, 1831) affected by white-nose syndrome. Evol Appl 2024; 17:e13748. [PMID: 39310794 PMCID: PMC11413065 DOI: 10.1111/eva.13748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/06/2023] [Revised: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 09/25/2024] Open
Abstract
Genome-wide scans for selection have become a popular tool for investigating evolutionary responses in wildlife to emerging diseases. However, genome scans are susceptible to false positives and do little to demonstrate specific mechanisms by which loci impact survival. Linking putatively resistant genotypes to observable phenotypes increases confidence in genome scan results and provides evidence of survival mechanisms that can guide conservation and management efforts. Here we used an expression quantitative trait loci (eQTL) analysis to uncover relationships between gene expression and alleles associated with the survival of little brown bats (Myotis lucifugus) despite infection with the causative agent of white-nose syndrome. We found that 25 of the 63 single-nucleotide polymorphisms (SNPs) associated with survival were related to gene expression in wing tissue. The differentially expressed genes have functional annotations associated with the innate immune system, metabolism, circadian rhythms, and the cellular response to stress. In addition, we observed differential expression of multiple genes with survival implications related to loci in linkage disequilibrium with focal SNPs. Together, these findings support the selective function of these loci and suggest that part of the mechanism driving survival may be the alteration of immune and other responses in epithelial tissue.
Collapse
Affiliation(s)
- Robert Kwait
- Department of Ecology, Evolution and Natural ResourcesRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| | - Malin L. Pinsky
- Department of Ecology, Evolution and Natural ResourcesRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
- Department of Ecology and Evolutionary BiologyUniversity of California Santa CruzSanta CruzCaliforniaUSA
| | | | - Evan A. Eskew
- Institute for Interdisciplinary Data SciencesUniversity of IdahoMoscowIdahoUSA
| | - Kathleen Kerwin
- Department of Ecology, Evolution and Natural ResourcesRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| | - Brooke Maslo
- Department of Ecology, Evolution and Natural ResourcesRutgers, The State University of New JerseyNew BrunswickNew JerseyUSA
| |
Collapse
|
47
|
Wright CF, Sharp LN, Jackson L, Murray A, Ware JS, MacArthur DG, Rehm HL, Patel KA, Weedon MN. Guidance for estimating penetrance of monogenic disease-causing variants in population cohorts. Nat Genet 2024; 56:1772-1779. [PMID: 39075210 DOI: 10.1038/s41588-024-01842-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/15/2023] [Accepted: 06/24/2024] [Indexed: 07/31/2024]
Abstract
Penetrance is the probability that an individual with a pathogenic genetic variant develops a specific disease. Knowing the penetrance of variants for monogenic disorders is important for counseling of individuals. Until recently, estimates of penetrance have largely relied on affected individuals and their at-risk family members being clinically referred for genetic testing, a 'phenotype-first' approach. This approach substantially overestimates the penetrance of variants because of ascertainment bias. The recent availability of whole-genome sequencing data in individuals from very-large-scale population-based cohorts now allows 'genotype-first' estimates of penetrance for many conditions. Although this type of population-based study can underestimate penetrance owing to recruitment biases, it provides more accurate estimates of penetrance for secondary or incidental findings. Here, we provide guidance for the conduct of penetrance studies to ensure that robust genotypes and phenotypes are used to accurately estimate penetrance of variants and groups of similarly annotated variants from population-based studies.
Collapse
Affiliation(s)
- Caroline F Wright
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK.
| | - Luke N Sharp
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK
| | - Leigh Jackson
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK
| | - Anna Murray
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK
| | - James S Ware
- National Heart and Lung Institute and MRC Laboratory of Medical Sciences, Imperial College London, London, UK
- Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Daniel G MacArthur
- Centre for Population Genomics, Garvan Institute of Medical Research and UNSW Sydney, Sydney, New South Wales, Australia
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Heidi L Rehm
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kashyap A Patel
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK
| | - Michael N Weedon
- Department of Clinical and Biomedical Sciences, Medical School, University of Exeter, Exeter, UK.
| |
Collapse
|
48
|
Guo H, Urban AE, Wong WH. Prioritizing disease-related rare variants by integrating gene expression data. PLoS Genet 2024; 20:e1011412. [PMID: 39348415 PMCID: PMC11466430 DOI: 10.1371/journal.pgen.1011412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/16/2024] [Revised: 10/10/2024] [Accepted: 08/29/2024] [Indexed: 10/02/2024] Open
Abstract
Rare variants, comprising the vast majority of human genetic variations, are likely to have more deleterious impact in the context of human diseases compared to common variants. Here we present carrier statistic, a statistical framework to prioritize disease-related rare variants by integrating gene expression data. By quantifying the impact of rare variants on gene expression, carrier statistic can prioritize those rare variants that have large functional consequence in the patients. Through simulation studies and analyzing real multi-omics dataset, we demonstrated that carrier statistic is applicable in studies with limited sample size (a few hundreds) and achieves substantially higher sensitivity than existing rare variants association methods. Application to Alzheimer's disease reveals 16 rare variants within 15 genes with extreme carrier statistics. We also found strong excess of rare variants among the top prioritized genes in patients compared to that in healthy individuals. The carrier statistic method can be applied to various rare variant types and is adaptable to other omics data modalities, offering a powerful tool for investigating the molecular mechanisms underlying complex diseases.
Collapse
Affiliation(s)
- Hanmin Guo
- Department of Statistics, Stanford University, Stanford, California, United States of America
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
| | - Alexander Eckehart Urban
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, California, United States of America
- Department of Genetics, Stanford University School of Medicine, Stanford, California, United States of America
| | - Wing Hung Wong
- Department of Statistics, Stanford University, Stanford, California, United States of America
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
49
|
Rivera NV. Big data in sarcoidosis. Curr Opin Pulm Med 2024; 30:561-569. [PMID: 38967053 PMCID: PMC11309342 DOI: 10.1097/mcp.0000000000001102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 07/06/2024]
Abstract
PURPOSE OF REVIEW This review provides an overview of recent advancements in sarcoidosis research, focusing on collaborative networks, phenotype characterization, and molecular studies. It highlights the importance of collaborative efforts, phenotype characterization, and the integration of multilevel molecular data for advancing sarcoidosis research and paving the way toward personalized medicine. RECENT FINDINGS Sarcoidosis exhibits heterogeneous clinical manifestations influenced by various factors. Efforts to define sarcoidosis endophenotypes show promise, while technological advancements enable extensive molecular data generation. Collaborative networks and biobanks facilitate large-scale studies, enhancing biomarker discovery and therapeutic protocols. SUMMARY Sarcoidosis presents a complex challenge due to its unknown cause and heterogeneous clinical manifestations. Collaborative networks, comprehensive phenotype delineation, and the utilization of cutting-edge technologies are essential for advancing our understanding of sarcoidosis biology and developing personalized medicine approaches. Leveraging large-scale epidemiological resources and biobanks and integrating multilevel molecular data offer promising avenues for unraveling the disease's heterogeneity and improving patient outcomes.
Collapse
Affiliation(s)
- Natalia V Rivera
- Division of Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
50
|
Jensen M, Smolen C, Tyryshkina A, Pizzo L, Banerjee D, Oetjens M, Shimelis H, Taylor CM, Pounraja VK, Song H, Rohan L, Huber E, El Khattabi L, van de Laar I, Tadros R, Bezzina C, van Slegtenhorst M, Kammeraad J, Prontera P, Caberg JH, Fraser H, Banka S, Van Dijck A, Schwartz C, Voorhoeve E, Callier P, Mosca-Boidron AL, Marle N, Lefebvre M, Pope K, Snell P, Boys A, Lockhart PJ, Ashfaq M, McCready E, Nowacyzk M, Castiglia L, Galesi O, Avola E, Mattina T, Fichera M, Bruccheri MG, Mandarà GML, Mari F, Privitera F, Longo I, Curró A, Renieri A, Keren B, Charles P, Cuinat S, Nizon M, Pichon O, Bénéteau C, Stoeva R, Martin-Coignard D, Blesson S, Le Caignec C, Mercier S, Vincent M, Martin C, Mannik K, Reymond A, Faivre L, Sistermans E, Kooy RF, Amor DJ, Romano C, Andrieux J, Girirajan S. Genetic modifiers and ascertainment drive variable expressivity of complex disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.27.24312158. [PMID: 39252907 PMCID: PMC11383473 DOI: 10.1101/2024.08.27.24312158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Academic Contribution Register] [Indexed: 09/11/2024]
Abstract
Variable expressivity of disease-associated variants implies a role for secondary variants that modify clinical features. We assessed the effects of modifier variants towards clinical outcomes of 2,252 individuals with primary variants. Among 132 families with the 16p12.1 deletion, distinct rare and common variant classes conferred risk for specific developmental features, including short tandem repeats for neurological defects and SNVs for microcephaly, while additional disease-associated variants conferred multiple genetic diagnoses. Within disease and population cohorts of 773 individuals with the 16p12.1 deletion, we found opposing effects of secondary variants towards clinical features across ascertainments. Additional analysis of 1,479 probands with other primary variants, such as 16p11.2 deletion and CHD8 variants, and 1,084 without primary variants, showed that phenotypic associations differed by primary variant context and were influenced by synergistic interactions between primary and secondary variants. Our study provides a paradigm to dissect the genomic architecture of complex disorders towards personalized treatment.
Collapse
Affiliation(s)
- Matthew Jensen
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
| | - Corrine Smolen
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
| | - Anastasia Tyryshkina
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Lucilla Pizzo
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Deepro Banerjee
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Matthew Oetjens
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Hermela Shimelis
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Cora M. Taylor
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Vijay Kumar Pounraja
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
| | - Hyebin Song
- Department of Statistics, Pennsylvania State University, University Park, PA 16802, USA
| | - Laura Rohan
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Emily Huber
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Laila El Khattabi
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, CARPEM, Paris, France
| | - Ingrid van de Laar
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Rafik Tadros
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Connie Bezzina
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marjon van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Janneke Kammeraad
- Department of Clinical Genetics, Erasmus MC, Univ. Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Paolo Prontera
- Medical Genetics Unit, Hospital Santa Maria della Misericordia, Perugia, Italy
| | - Jean-Hubert Caberg
- Centre Hospitalier Universitaire de Liège. Domaine Universitaire du Sart Tilman, Liège, Belgium
| | - Harry Fraser
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Siddhartha Banka
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, St. Mary’s Hospital, Central Manchester University Hospitals, NHS Foundation Trust Manchester Academic Health Sciences Centre, Manchester, UK
| | - Anke Van Dijck
- Department of Medical Genetics, University and University Hospital Antwerp, Antwerp, Belgium
| | | | - Els Voorhoeve
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Patrick Callier
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
| | - Anne-Laure Mosca-Boidron
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
| | - Nathalie Marle
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
| | - Mathilde Lefebvre
- Laboratoire de Genetique Chromosomique et Moleculaire, CHU Dijon, France
| | - Kate Pope
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Penny Snell
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Amber Boys
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
| | - Paul J. Lockhart
- Bruce Lefroy Centre, Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Myla Ashfaq
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Elizabeth McCready
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Margaret Nowacyzk
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Lucia Castiglia
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Ornella Galesi
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Emanuela Avola
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Teresa Mattina
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | - Marco Fichera
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
- Section of Clinical Biochemistry and Medical Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania School of Medicine, Catania, Italy
| | - Maria Grazia Bruccheri
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
| | | | - Francesca Mari
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Flavia Privitera
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Longo
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Aurora Curró
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Renieri
- Laboratory of Clinical Molecular Genetics and Cytogenetics, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Boris Keren
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 75019 Paris, France
| | - Perrine Charles
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, 75019 Paris, France
| | | | | | | | | | - Radka Stoeva
- CHU Nantes, Medical Genetics Department, Nantes, France
| | | | - Sophia Blesson
- Department of Genetics, Bretonneau University Hospital, Tours, France
| | - Cedric Le Caignec
- CHU Toulouse, Department of Medical Genetics, Toulouse, France
- Toulouse Neuro Imaging, Center, Inserm, UPS, Université de Toulouse, Toulouse, France
| | - Sandra Mercier
- Department of Genetics, Bretonneau University Hospital, Tours, France
| | - Marie Vincent
- Department of Genetics, Bretonneau University Hospital, Tours, France
| | - Christa Martin
- Autism & Developmental Medicine Institute, Geisinger, Lewisburg, PA 17837, USA
| | - Katrin Mannik
- Institute of Genomics, University of Tartu, Estonia
- Health2030 Genome Center, Fondation Campus Biotech, Geneva, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Switzerland
| | - Laurence Faivre
- Center for Rare Diseases and Reference Developmental Anomalies and Malformation Syndromes, CHU Dijon, Dijon, France
- Laboratoire de Genetique Chromosomique et Moleculaire, CHU Dijon, France
| | - Erik Sistermans
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - R. Frank Kooy
- Department of Medical Genetics, University and University Hospital Antwerp, Antwerp, Belgium
| | - David J. Amor
- Department of Clinical Genetics, Amsterdam UMC, Amsterdam, The Netherlands
| | - Corrado Romano
- Research Unit of Rare Diseases and Neurodevelopmental Disorders, Oasi Research Institute-IRCCS, Troina, Italy
- Section of Clinical Biochemistry and Medical Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania School of Medicine, Catania, Italy
| | - Joris Andrieux
- Institut de Genetique Medicale, Hopital Jeanne de Flandre, CHRU de Lille, Lille, France
| | - Santhosh Girirajan
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA 16802, USA
- Bioinformatics and Genomics Graduate program, Pennsylvania State University, University Park, PA 16802, USA
- Department of Anthropology, Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|