1
|
Cui Y, Chen Y, Hu M, Zhou H, Guo J, Wang Q, Xu Z, Chen L, Zhang W, Tang S. Bidirectional Mendelian randomization and colocalization analysis of gut microbiota on lipid profile. Comput Biol Chem 2025; 117:108422. [PMID: 40080991 DOI: 10.1016/j.compbiolchem.2025.108422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 01/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
The gut microbiota plays a crucial role in human health, but its impact on lipid metabolism remains unclear. Understanding the causal relationship between gut bacteria and lipid profiles is essential for developing strategies to prevent and treat dyslipidemia and cardiovascular diseases. This study aimed to assess this relationship using two-sample Mendelian randomization (MR). Data for both exposure and outcomes were obtained from the IEU-GWAS database, with lipid profile data sourced from a publication. Genome-wide significant single nucleotide polymorphisms (SNPs), which were independent of outcome factors but correlated with exposure variables, were identified as instrumental variables. Several MR methods, including weighted analysis, maximum likelihood, inverse variance weighting (IVW), MR-Egger, and weighted median, were applied. Colocalization analysis further validated the findings. The analysis revealed microbial groups with causal relationships to ApoA1, ApoB, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, total cholesterol, and triglycerides. Reverse MR and colocalization analysis provided additional confirmation of these results. This study offers new evidence of the causal link between gut microbiota and lipid profiles, providing insights for improving lipid profiles and reducing cardiovascular disease risk.
Collapse
Affiliation(s)
- Yu Cui
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China
| | - Yanzhu Chen
- Operating Room 1 Area, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Mengting Hu
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China
| | - He Zhou
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Jiarui Guo
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Qijia Wang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Zaihua Xu
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Liyun Chen
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Research Center of Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Wancong Zhang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China.
| | - Shijie Tang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China.
| |
Collapse
|
2
|
Afghan TS, Khan SN, Awan FM, Obaid A, Basri R, Ullah A, Khan S, Naz A, Ullah K, Jabbar A. An integrated approach for genetic risk profiling of typhoid, tuberculosis, and cholera in local population of tehsil Haripur. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2025; 131:105756. [PMID: 40339732 DOI: 10.1016/j.meegid.2025.105756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/10/2025]
Abstract
Despite notable progress in public health throughout the 21st century, infectious diseases like tuberculosis, typhoid, and cholera remain serious threats to global health, particularly in high-risk regions. Understanding the genetic factors that influence susceptibility and resistance to these diseases is essential for developing more effective strategies for their prevention and treatment. This study investigates the genetic variations associated with these infectious diseases with a focus on regions where these diseases are most prevalent. The aim of this study is to identify genetic variants that may influence susceptibility to tuberculosis, typhoid, and cholera. A thorough analysis of genetic variants associated with susceptibility and resistance to tuberculosis, typhoid, and cholera was conducted. Using publicly available genetic data from various ethnic groups. The allele frequency of the prioritized variants was calculated to assess their distribution within the different populations, including Middle Eastern, Ashkenazi Jewish, European (Non-Finnish), Latino/Admixed American, South Asian, East Asian, European (Finnish), North Asian, Southeast Asian, African American, and Swedish populations. The variants of the IL1β gene exhibiting the highest allele frequencies in the South Asian population were identified and subsequently examined within the local population. Specifically, the variants rs1143627 and rs1143629, which demonstrate the highest allele frequencies and are associated with typhoid, tuberculosis, and cholera, were subjected to detailed analysis. To determine their distribution and potential impact on disease susceptibility. In the local population, statistical analysis of the available sample revealed allele frequencies of 0.1128 % for IL1β (rs1143627) and 0.18 % for IL1β (rs1143629). Furthermore, these findings revealed that certain genetic profiles may play a role in the population's overall response to infectious diseases such as tuberculosis, typhoid, and cholera. This research has the potential to guide future public health strategies for more effective management and prevention of these diseases.
Collapse
Affiliation(s)
- Tahira Sher Afghan
- Department of Medical Lab Technology, The University of Haripur (UOH), Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Sadiq Noor Khan
- Department of Medical Lab Technology, The University of Haripur (UOH), Haripur, Khyber Pakhtunkhwa, Pakistan.
| | - Faryal Mehwish Awan
- Department of Medical Lab Technology, The University of Haripur (UOH), Haripur, Khyber Pakhtunkhwa, Pakistan.
| | - Ayesha Obaid
- Department of Medical Lab Technology, The University of Haripur (UOH), Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Rabea Basri
- Department of Medical Lab Technology, The University of Haripur (UOH), Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Amin Ullah
- Department of Medical Lab Technology, The University of Haripur (UOH), Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Saira Khan
- Department of Medical Lab Technology, The University of Haripur (UOH), Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Anam Naz
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore (UOL), Lahore, Pakistan
| | - Kamran Ullah
- Department of Biology, The University of Haripur (UOH), Haripur, Khyber Pakhtunkhwa, Pakistan
| | - Abdul Jabbar
- Department of Medical Lab Technology, The University of Haripur (UOH), Haripur, Khyber Pakhtunkhwa, Pakistan
| |
Collapse
|
3
|
Su T, Xia Y. A quantitative comparison of the deleteriousness of missense and nonsense mutations using the structurally resolved human protein interactome. Protein Sci 2025; 34:e70155. [PMID: 40384578 PMCID: PMC12086521 DOI: 10.1002/pro.70155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/02/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
The complex genotype-to-phenotype relationships in Mendelian diseases can be elucidated by mutation-induced disturbances to the networks of molecular interactions (interactomes) in human cells. Missense and nonsense mutations cause distinct perturbations within the human protein interactome, leading to functional and phenotypic effects with varying degrees of severity. Here, we structurally resolve the human protein interactome at atomic-level resolutions and perform structural and thermodynamic calculations to assess the biophysical implications of these mutations. We focus on a specific type of missense mutation, known as "quasi-null" mutations, which destabilize proteins and cause similar functional consequences (node removal) to nonsense mutations. We propose a "fold difference" quantification of deleteriousness, which measures the ratio between the fractions of node-removal mutations in datasets of Mendelian disease-causing and non-pathogenic mutations. We estimate the fold differences of node-removal mutations to range from 3 (for quasi-null mutations with folding ΔΔG ≥2 kcal/mol) to 20 (for nonsense mutations). We observe a strong positive correlation between biophysical destabilization and phenotypic deleteriousness, demonstrating that the deleteriousness of quasi-null mutations spans a continuous spectrum, with nonsense mutations at the extreme (highly deleterious) end. Our findings substantiate the disparity in phenotypic severity between missense and nonsense mutations and suggest that mutation-induced protein destabilization is indicative of the phenotypic outcomes of missense mutations. Our analyses of node-removal mutations allow for the potential identification of proteins whose removal or destabilization lead to harmful phenotypes, enabling the development of targeted therapeutic approaches, and enhancing comprehension of the intricate mechanisms governing genotype-to-phenotype relationships in clinically relevant diseases.
Collapse
Affiliation(s)
- Ting‐Yi Su
- Graduate Program in Quantitative Life SciencesMcGill UniversityMontréalQuébecCanada
| | - Yu Xia
- Graduate Program in Quantitative Life SciencesMcGill UniversityMontréalQuébecCanada
- Department of BioengineeringMcGill UniversityMontréalQuébecCanada
| |
Collapse
|
4
|
Scheinfeldt L, Kusic D, Gaedigk A, Turner AJ, Moyer AM, Pratt VM, Kalman LV. New Resources to Identify Characterized DNA Reference Materials for Pharmacogenetic (PGx) and Human Leukocyte Antigen (HLA) Testing: The Genetic Testing Reference Material (GeT-RM) Program PGx Search Tool and GeT-RM Consolidated PGx and HLA Table. J Mol Diagn 2025; 27:457-464. [PMID: 40122159 PMCID: PMC12103986 DOI: 10.1016/j.jmoldx.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/10/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Regulations, accreditation standards, and professional guidance require laboratories to use reference materials for assay development, validation, quality control, and proficiency testing of clinical genetic tests. There are, however, few publicly available reference materials for most genetic tests. To address this issue, the CDC's Genetic Testing Reference Material Program (GeT-RM), the Coriell Institute for Medical Research, and the genetic testing community have conducted 19 studies, including nine for pharmacogenetic (PGx) and human leukocyte antigen (HLA) testing, to generate characterized, renewable, and publicly available DNA samples for use as reference materials. Because new PGx alleles are frequently identified, and allele designations change over time, many samples were reanalyzed for the same gene(s) in subsequent GeT-RM studies. These studies used more comprehensive and sensitive methods and panels that examined additional single-nucleotide variants and/or star alleles to expand and update the consensus genotypes. Up-to-date information is available in two newly established resources: the GeT-RM Consolidated PGx and HLA Table and the GeT-RM PGx Search Tool. These resources contain all available PGx and HLA genotypes for 363 publicly available samples characterized during nine GeT-RM PGx or HLA studies for 34 genes/loci in a consolidated and searchable format.
Collapse
Affiliation(s)
| | - Dara Kusic
- Coriell Institute for Medical Research, Camden, New Jersey
| | - Andrea Gaedigk
- Children's Mercy Research Institute, Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, University of Missouri-Kansas City School of Medicine, Kansas City, Missouri
| | - Amy J Turner
- RPRD Diagnostics and the Department of Pediatrics, Section on Genomic Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Ann M Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Victoria M Pratt
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana; Agena Bioscience, San Diego, California
| | - Lisa V Kalman
- Division of Laboratory Systems, Centers for Disease Control and Prevention, Atlanta, Georgia.
| |
Collapse
|
5
|
Berthold N, MacDermod CM, Thornton LM, Parker R, Morales SAC, Hog L, Kennedy HL, Guintivano J, Sullivan PF, Crowley JJ, Johnson JS, Birgegård A, Fundín BT, Frans E, Xu J, Ngāti Pūkenga MP, Miller AL, Aguilar MV, Barakat S, Abdulkadir M, White JP, Larsen JT, Trujillo E, Winterman B, Zhang R, Lawson R, Wonderlich S, Wonderlich J, Schaefer LM, Mehler PS, Oakes J, Foster M, Gaudiani J, Vacuán ETC, Compte EJ, Petersen LV, Yilmaz Z, Micali N, Jordan J, Kennedy MA, Maguire S, Huckins LM, Lu Y, Dinkler L, Martin NG, Bulik CM. The Eating Disorders Genetics Initiative 2 (EDGI2): study protocol. BMC Psychiatry 2025; 25:532. [PMID: 40419993 DOI: 10.1186/s12888-025-06777-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND The Eating Disorders Genetics Initiative 2 (EDGI2) is designed to explore the role of genes and environment in anorexia nervosa, bulimia nervosa, binge-eating disorder, and avoidant/restrictive food intake disorder (ARFID) with a focus on broad population representation and severe and/or longstanding illness. METHODS A total of 20,000 new participants (18,700 cases and 1,300 controls) will be ascertained from the United States (US), Mexico (MX), Australia (AU), Aotearoa New Zealand (NZ), Sweden (SE), and Denmark (DK). Comprehensive phenotyping and genotyping will be performed for participants in US, MX, AU, NZ, and SE using the EDGI2 questionnaire battery and participant saliva samples. In DK, case identification and genotyping will be through the National Patient Register and bloodspots archived near birth. Case-control and case-case genome-wide association studies will be conducted within EDGI2 and enhanced via meta-analysis with external data from the Eating Disorders Working Group of the Psychiatric Genomics Consortium (PGC-ED). Additional analyses will explore genetic correlations between eating disorders (EDs) and other psychiatric and metabolic traits, calculate polygenic risk scores (PRS), and leverage functional biology to evaluate clinical outcomes. Moreover, analyzing PRS for patient stratification and linking identified risk loci to clinically relevant phenotypes highlight the potential of EDGI2 for clinical translation. DISCUSSION EDGI2 is a global expansion of the EDGI study to increase sample size, increase participant representation across multiple ancestral backgrounds, and to include ARFID. ED genetics research has historically lagged behind other psychiatric disorders, and EDGI2 is designed to rapidly advance the study of the genetics of the major EDs. Exploring EDs at both the diagnostic level and the symptom level will provide an unprecedented look at the genetic architecture underlying EDs. TRIAL REGISTRATION EDGI2 is a registered clinical trial: clinicaltrials.gov NCT06594913. https://clinicaltrials.gov/study/NCT06594913 (posted September 19, 2024).
Collapse
Affiliation(s)
- Natasha Berthold
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
- School of Human Sciences, University of Western Australia, Crawley, WA, 6009, Australia
- Perron Research Institute, Nedlands, WA, 6009, Australia
| | - Casey M MacDermod
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Laura M Thornton
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Richard Parker
- QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Locked Bag 2000, Brisbane, QLD, 4029, Australia
| | - Shantal Anid Cortés Morales
- Research Department, Comenzar de Nuevo, Monterrey, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud - , Monterrey, Mexico
| | - Liv Hog
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Hannah L Kennedy
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Jerry Guintivano
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Patrick F Sullivan
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - James J Crowley
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Jessica S Johnson
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Andreas Birgegård
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Bengt T Fundín
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Emma Frans
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Jiayi Xu
- Division of Molecular Psychiatry, Department of Psychiatry, Yale University, New Haven, CT, USA
| | | | - Allison L Miller
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Mariana Valdez Aguilar
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
| | - Sarah Barakat
- Insideout Institute for Eating Disorders, The University of Sydney, Sydney, Australia
| | - Mohamed Abdulkadir
- Department of Public Health, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Jennifer P White
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
- Department of Psychology, University of Albany, State University of New York, Albany, NY, USA
| | - Janne T Larsen
- Department of Public Health, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Elsie Trujillo
- Research Department, Comenzar de Nuevo, Monterrey, Mexico
| | | | - Ruyue Zhang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Rachel Lawson
- South Island Eating Disorders Service, Health NZ Te Whatu Ora, Christchurch, New Zealand
| | - Stephen Wonderlich
- Center for Biobehavioral Research, Sanford Health, Fargo, ND, USA
- Department of Psychiatry and Behavioral Science, University of North Dakota School of Medicine and Health Sciences, Fargo, ND, USA
| | | | | | - Philip S Mehler
- ACUTE Center for Eating Disorders and Severe Malnutrition, Denver Health and Hospital Authority, Denver, CO, USA
| | - Judy Oakes
- ACUTE Center for Eating Disorders and Severe Malnutrition, Denver Health and Hospital Authority, Denver, CO, USA
| | - Marina Foster
- ACUTE Center for Eating Disorders and Severe Malnutrition, Denver Health and Hospital Authority, Denver, CO, USA
| | | | - Eva Trujillo Chi Vacuán
- Research Department, Comenzar de Nuevo, Monterrey, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud - , Monterrey, Mexico
| | - Emilio J Compte
- Research Department, Comenzar de Nuevo, Monterrey, Mexico
- Eating Behavior Research Center, School of Psychology, Universidad Adolfo Ibáñez, Santiago, Chile
| | - Liselotte V Petersen
- Department of Public Health, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
| | - Zeynep Yilmaz
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA
- Department of Public Health, National Centre for Register-Based Research, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Nadia Micali
- Center for Eating and Feeding Disorders Research, Mental Health Services of the Capital Region of Denmark, Psychiatric Centre Ballerup, Copenhagen, Denmark
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Jennifer Jordan
- Department of Psychological Medicine, University of Otago, Christchurch, New Zealand
- Health NZ - Te Whatu Ora, Christchurch, New Zealand
| | - Martin A Kennedy
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud - , Monterrey, Mexico
| | - Sarah Maguire
- Insideout Institute for Eating Disorders, The University of Sydney, Sydney, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Laura M Huckins
- Division of Molecular Psychiatry, Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Yi Lu
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Lisa Dinkler
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Nicholas G Martin
- QIMR Berghofer Medical Research Institute, Royal Brisbane Hospital, Locked Bag 2000, Brisbane, QLD, 4029, Australia
| | - Cynthia M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, #7160, 101 Manning Drive, Chapel Hill, CBNC, 27599 - 7160, USA.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, 171 77, Stockholm, Sweden.
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
6
|
Hong SC, Muyas F, Cortés-Ciriano I, Hormoz S. scAI-SNP: a method for inferring ancestry from single-cell data. BMC METHODS 2025; 2:10. [PMID: 40401145 PMCID: PMC12089154 DOI: 10.1186/s44330-025-00029-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 05/01/2025] [Indexed: 05/23/2025]
Abstract
Background Collaborative efforts, such as the Human Cell Atlas, are rapidly accumulating large amounts of single-cell data. To ensure that single-cell atlases are representative of human genetic diversity, we need to determine the ancestry of the donors from whom single-cell data are generated. Self-reporting of race and ethnicity, although important, can be biased and is not always available for the datasets already collected. Methods Here, we introduce scAI-SNP, a tool to infer ancestry directly from single-cell genomics data. To train scAI-SNP, we identified 4.5 million ancestry-informative single-nucleotide polymorphisms (SNPs) in the 1000 Genomes Project dataset across 3201 individuals from 26 population groups. For a query single-cell dataset, scAI-SNP uses these ancestry-informative SNPs to compute the contribution of each of the 26 population groups to the ancestry of the donor from whom the cells were obtained. Results Using diverse single-cell datasets with matched whole-genome sequencing data, we show that scAI-SNP is robust to the sparsity of single-cell data, can accurately and consistently infer ancestry from samples derived from diverse types of tissues and cancer cells, and can be applied to different modalities of single-cell profiling assays, such as single-cell RNA-seq and single-cell ATAC-seq. Discussion Finally, we argue that ensuring that single-cell atlases represent diverse ancestry, ideally alongside race and ethnicity, is ultimately important for improved and equitable health outcomes by accounting for human diversity. Supplementary Information The online version contains supplementary material available at 10.1186/s44330-025-00029-4.
Collapse
Affiliation(s)
- Sung Chul Hong
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215 USA
| | - Francesc Muyas
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge, CB10 1SD UK
| | - Isidro Cortés-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge, CB10 1SD UK
| | - Sahand Hormoz
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02215 USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115 USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| |
Collapse
|
7
|
Talenti A, Wilkinson T, Morrison LJ, Prendergast JGD. The evolution and convergence of mutation spectra across mammals. Commun Biol 2025; 8:763. [PMID: 40379828 DOI: 10.1038/s42003-025-08181-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
Despite the key role genetic mutations play in shaping phenotypic differences between species, little is currently known about the evolution of germline mutation spectra across mammals. Domesticated species are likely particularly interesting case studies because of their high mutation rates and complex evolutionary histories, which can span multiple founding events and genetic bottlenecks. Here we have developed a new reusable workflow, nSPECTRa, that can undertake the key steps in characterising mutation spectra, from determining ancestral alleles to characterising multiple forms of variation. We apply nSPECTRa to seven species, including several that have undergone domestication, and highlight how nSPECTRa can provide important insights into mutation rate evolution. While mutation spectra most often show marked differences between species and even breeds, certain mutation types have risen to a high frequency in subpopulations of different species, indicative of convergent evolution in mutation rates. This includes the previously characterized TCC- > TTC change enriched among European humans, which is also enriched among East Asian cattle. We show Indicine cattle are particularly interesting examples of how different mutation spectra segregate within a population and subsequently spread across the globe. Together, this work has important implications for understanding the control and evolution of mammalian mutation rates.
Collapse
Affiliation(s)
- Andrea Talenti
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, UK.
| | - Toby Wilkinson
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - Liam J Morrison
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, UK
| | - James G D Prendergast
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian, UK.
| |
Collapse
|
8
|
Lerga-Jaso J, Novković B, Unnikrishnan D, Bamunusinghe V, Hatorangan MR, Manson C, Pedersen H, Osama A, Terpolovsky A, Bohn S, De Marino A, Mahmoud AA, Bircan KO, Khan U, Grabherr MG, Yazdi PG. Tracing human genetic histories and natural selection with precise local ancestry inference. Nat Commun 2025; 16:4576. [PMID: 40379651 PMCID: PMC12084304 DOI: 10.1038/s41467-025-59936-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
Local ancestry inference is crucial for unraveling demographic histories, discovering selection signals, and including admixed individuals in genomic studies for improved equity and portability. To date, the precision and resolution of local ancestry inference were limited by technical and dataset issues. To address them, we present Orchestra, a model we train on over 10,000 single-origin individuals from 35 worldwide populations that demonstrates superior accuracy in benchmarking analyzes. We employ Orchestra to shed light on the demographic history of Latin Americans, finding trace ancestries supported by historical records. We then deploy it to offer insight on the debated Ashkenazi Jewish origins, highlighting their South European heritage. Finally, Orchestra enables us to map selection signatures, identifying trace Scandinavian ancestry in British samples and unveiling an immune-rich region linked to respiratory infections passed down from the Viking conquests. Our work significantly advances the field of local ancestry inference, highlighting its use in admixed populations.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Alex Osama
- Research & Development, Omicsedge, Miami, FL, USA
| | | | - Sandra Bohn
- Research & Development, Omicsedge, Miami, FL, USA
| | | | | | | | - Umar Khan
- Research & Development, Omicsedge, Miami, FL, USA
| | | | - Puya G Yazdi
- Research & Development, Omicsedge, Miami, FL, USA.
| |
Collapse
|
9
|
Xiong H, Shen P, Luo Q, Zhang L, Li B, Ding Z, Wang L. Elucidating the Genetic Underpinnings of Human Musculoskeletal System Aging Through Genomic Structural Equation Modeling. Clin Genet 2025. [PMID: 40369705 DOI: 10.1111/cge.14766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/11/2025] [Accepted: 04/30/2025] [Indexed: 05/16/2025]
Abstract
The genetic architecture underlying traits related to Human Musculoskeletal System Aging (MSA) remains largely unexplored. In this study, we conducted a large-scale multivariate genome-wide association study (GWAS) of MSA utilizing Genomic Structural Equation Modeling (Genomic SEM). We estimated causal single nucleotide polymorphisms (SNPs) associated with independent variation and identified 14 genome-wide significant loci (mean.PP > 0.95). We employed multiple transcriptome-wide association methods to analyze tissue, cellular levels, and genomic elements, identifying loci with high relevance to MSA susceptibility, along with associated element information. Our research represents the first comprehensive delineation of the genetic architecture of Musculoskeletal System Aging through a GWAS of unmeasured phenotype.
Collapse
Affiliation(s)
- Hao Xiong
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Pan Shen
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Qinghua Luo
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
| | - Leichang Zhang
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Department of Anorectal Surgery, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Bo Li
- Medical Department, Sias University, Zhengzhou, China
| | - Zhaohui Ding
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Pulmonary Disease Department, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Lihua Wang
- Department of Clinical Medicine, Jiangxi University of Chinese Medicine, Nanchang, China
- Pulmonary Disease Department, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| |
Collapse
|
10
|
He Y, Zhang X, Peng MS, Li YC, Liu K, Zhang Y, Mao L, Guo Y, Ma Y, Zhou B, Zheng W, Yue T, Liao Y, Liang SA, Chen L, Zhang W, Chen X, Tang B, Yang X, Ye K, Gao S, Lu Y, Wang Y, Wan S, Hao R, Wang X, Mao Y, Dai S, Gao Z, Yang LQ, Guo J, Li J, Liu C, Wang J, Sovannary T, Bunnath L, Kampuansai J, Inta A, Srikummool M, Kutanan W, Ho HQ, Pham KD, Singthong S, Sochampa S, Kyaing UW, Pongamornkul W, Morlaeku C, Rattanakrajangsri K, Kong QP, Zhang YP, Su B. Genome diversity and signatures of natural selection in mainland Southeast Asia. Nature 2025:10.1038/s41586-025-08998-w. [PMID: 40369069 DOI: 10.1038/s41586-025-08998-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Mainland Southeast Asia (MSEA) has rich ethnic and cultural diversity with a population of nearly 300 million1,2. However, people from MSEA are underrepresented in the current human genomic databases. Here we present the SEA3K genome dataset (phase I), generated by deep short-read whole-genome sequencing of 3,023 individuals from 30 MSEA populations, and long-read whole-genome sequencing of 37 representative individuals. We identified 79.59 million small variants and 96,384 structural variants, among which 22.83 million small variants and 24,622 structural variants are unique to this dataset. We observed a high genetic heterogeneity across MSEA populations, reflected by the varied combinations of genetic components. We identified 44 genomic regions with strong signatures of Darwinian positive selection, covering 89 genes involved in varied physiological systems such as physical traits and immune response. Furthermore, we observed varied patterns of archaic Denisovan introgression in MSEA populations, supporting the proposal of at least two distinct instances of Denisovan admixture into modern humans in Asia3. We also detected genomic regions that suggest adaptive archaic introgressions in MSEA populations. The large number of novel genomic variants in MSEA populations highlight the necessity of studying regional populations that can help answer key questions related to prehistory, genetic adaptation and complex diseases.
Collapse
Affiliation(s)
- Yaoxi He
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Yunnan Key Laboratory of Integrative Anthropology, Kunming, China
| | - Xiaoming Zhang
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Yunnan Key Laboratory of Integrative Anthropology, Kunming, China
| | - Min-Sheng Peng
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu-Chun Li
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, China
- Kunming Key Laboratory of Healthy Aging Study, Kunming, China
| | - Kai Liu
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yu Zhang
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Leyan Mao
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yongbo Guo
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Yujie Ma
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bin Zhou
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Wangshan Zheng
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tian Yue
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yuwen Liao
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shen-Ao Liang
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Science, Fudan University, Shanghai, China
| | - Lu Chen
- State Key Laboratory of Genetic Engineering, Center for Evolutionary Biology, School of Life Science, Fudan University, Shanghai, China
| | - Weijie Zhang
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaoning Chen
- National Genomics Data Center, China National Center for Bioinformation, Beijing, China
| | - Bixia Tang
- National Genomics Data Center, China National Center for Bioinformation, Beijing, China
| | - Xiaofei Yang
- School of Computer Science and Technology, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
- Center for Mathematical Medical, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Kai Ye
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
- Center for Mathematical Medical, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
- Genome Institute, the First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
- Faculty of Science, Leiden University, Leiden, The Netherlands
| | - Shenghan Gao
- School of Computer Science and Technology, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Yurun Lu
- CEMS, NCMIS, HCMS, MADIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
| | - Yong Wang
- CEMS, NCMIS, HCMS, MADIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
| | - Shijie Wan
- School of Computer Science and Technology, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Rushan Hao
- School of Medicine, Yunnan University, Kunming, China
| | - Xuankai Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Yafei Mao
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
- Center for Genomic Research, International Institutes of Medicine, The Fourth Affiliated Hospital, Zhejiang University, Yiwu, China
| | - Shanshan Dai
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Zongliang Gao
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
- Kunming Key Laboratory of Healthy Aging Study, Kunming, China
| | - Li-Qin Yang
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Yunnan Key Laboratory of Integrative Anthropology, Kunming, China
- Kunming Key Laboratory of Healthy Aging Study, Kunming, China
| | - Jianxin Guo
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Jiangguo Li
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chao Liu
- Laboratory Animal Center, Kunming Institute of Zoology, the Chinese Academy of Sciences, Kunming, China
- National Resource Center for Non-Human Primates, Kunming, China
| | - Jianhua Wang
- Department of Anthropology, School of Sociology, Yunnan Minzu University, Kunming, China
| | - Tuot Sovannary
- Department of Geography and Land Management, Royal University of Phnom Penh, Phnom Penh, Cambodia
| | - Long Bunnath
- Department of Geography and Land Management, Royal University of Phnom Penh, Phnom Penh, Cambodia
| | - Jatupol Kampuansai
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
| | - Angkhana Inta
- Department of Biology, Faculty of Science, Chiang Mai University, Chiang Mai, Thailand
| | - Metawee Srikummool
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Wibhu Kutanan
- Department of Biology, Faculty of Science, Naresuan University, Phitsanulok, Thailand
| | - Huy Quang Ho
- Department of Immunology, Ha Noi Medical University, Ha Noi, Vietnam
| | - Khoa Dang Pham
- Department of Immunology, Ha Noi Medical University, Ha Noi, Vietnam
| | | | | | - U Win Kyaing
- Field School of Archaeology, Paukkhaung, Myanmar
| | - Wittaya Pongamornkul
- Queen Sirikit Botanic Garden (QSBG), The Botanical Garden Organization, Chiang Mai, Thailand
| | - Chutima Morlaeku
- Inter Mountain Peoples Education and Culture in Thailand Association (IMPECT), Sansai, Thailand
| | | | - Qing-Peng Kong
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, China.
- Kunming Key Laboratory of Healthy Aging Study, Kunming, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming, China.
- University of Chinese Academy of Sciences, Beijing, China.
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, School of Life Sciences, Yunnan University, Kunming, China.
| | - Bing Su
- State Key Laboratory of Genetic Evolution and Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China.
- Yunnan Key Laboratory of Integrative Anthropology, Kunming, China.
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
11
|
Acosta-Uribe J, Escudero SDP, Cochran JN, Taylor JW, Castruita A, Jonson C, Barinaga EA, Roberts K, Levine AR, George DS, AvilaFunes JA, Behrens MI, Bruno MA, Brusco LI, Custodio N, Duran-Aniotz C, Lopera F, Matallana DL, Slachevsky A, Takada LT, Zapata-Restrepo LM, Durón-Reyes DE, de Paula França Resende E, Gelvez N, Gómez LF, Godoy ME, Maito MA, Avandel S, Miller BL, Nalls MA, Leonard H, Vitale D, Bandres-Ciga S, Koretsky MJ, Singleton AB, Pantazis CB, Valcour V, Ibañez A, Kosik KS, Yokoyama JS, Bistue MB. Genetic Contributions to Alzheimer's Disease and Frontotemporal Dementia in Admixed Latin American Populations. RESEARCH SQUARE 2025:rs.3.rs-5462510. [PMID: 40386425 PMCID: PMC12083672 DOI: 10.21203/rs.3.rs-5462510/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Latin America's diverse genetic landscape provides a unique opportunity to study Alzheimer's disease (AD) and frontotemporal dementia (FTD). The Multi-Partner Consortium to Expand Dementia Research in Latin America (ReDLat) recruited 2,162 participants with AD, FTD, or as healthy controls from six countries: Argentina, Brazil, Chile, Colombia, Mexico, and Peru. Participants underwent genomic sequencing and population structure analyses were conducted using Principal Component Analysis and ADMIXTURE. The study revealed a predominant mix of American, African, and European ancestries, with an additional East Asian component in Brazil. Variant curation identified 17 pathogenic variants, pathogenic C9orf72 expansion, and 44 variants of uncertain significance. Seventy families showed autosomal dominant inheritance, with 48 affected by AD and 22 by FTD. This represents the first large-scale genetic study of AD and FTD in Latin America, highlighting the need to consider diverse ancestries, social determinants of health, and cultural factors when assessing genetic risk for neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Leonel T Takada
- Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo
| | | | | | | | | | | | | | | | | | | | | | | | - Dan Vitale
- Data Tecnica International (United States)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Mews MA, Naj AC, Griswold AJ, Below JE, Bush WS. Brain and blood transcriptome-wide association studies identify five novel genes associated with Alzheimer's disease. J Alzheimers Dis 2025; 105:228-244. [PMID: 40111921 DOI: 10.1177/13872877251326288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
BackgroundGenome-wide association studies (GWAS) have identified numerous genetic variants associated with Alzheimer's disease (AD), but their functional implications remain unclear. Transcriptome-wide association studies (TWAS) offer enhanced statistical power by analyzing genetic associations at the gene level rather than at the variant level, enabling assessment of how genetically-regulated gene expression influences AD risk. However, previous AD-TWAS have been limited by small expression quantitative trait loci (eQTL) reference datasets or reliance on AD-by-proxy phenotypes.ObjectiveTo perform the most powerful AD-TWAS to date using summary statistics from the largest available brain and blood cis-eQTL meta-analyses applied to the largest clinically-adjudicated AD GWAS.MethodsWe implemented the OTTERS TWAS pipeline to predict gene expression using the largest available cis-eQTL data from cortical brain tissue (MetaBrain; N = 2683) and blood (eQTLGen; N = 31,684), and then applied these models to AD-GWAS data (Cases = 21,982; Controls = 44,944).ResultsWe identified and validated five novel gene associations in cortical brain tissue (PRKAG1, C3orf62, LYSMD4, ZNF439, SLC11A2) and six genes proximal to known AD-related GWAS loci (Blood: MYBPC3; Brain: MTCH2, CYB561, MADD, PSMA5, ANXA11). Further, using causal eQTL fine-mapping, we generated sparse models that retained the strength of the AD-TWAS association for MTCH2, MADD, ZNF439, CYB561, and MYBPC3.ConclusionsOur comprehensive AD-TWAS discovered new gene associations and provided insights into the functional relevance of previously associated variants, which enables us to further understand the genetic architecture underlying AD risk.
Collapse
Affiliation(s)
- Makaela A Mews
- System Biology and Bioinformatics, Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Adam C Naj
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute and Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William S Bush
- Department of Population and Quantitative Health Sciences, Cleveland Institute for Computational Biology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
13
|
Malinga TV, Othman H, Paximadis M, Tiemessen CT, Ramsay M, Hazelhurst S, Twesigomwe D. Characterization of NAT, GST, and CYP2E1 Genetic Variation in Sub-Saharan African Populations: Implications for Treatment of Tuberculosis and Other Diseases. Clin Pharmacol Ther 2025; 117:1338-1357. [PMID: 39829327 PMCID: PMC11993289 DOI: 10.1002/cpt.3557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Tuberculosis (TB) is a major health burden in Africa. Although TB is treatable, anti-TB drugs are associated with adverse drug reactions (ADRs), which are partly attributed to pharmacogenetic variation. The distribution of star alleles (haplotypes) influencing anti-TB drug metabolism is unknown in many African populations. This presents challenges in implementing genotype-guided therapy in Africa to decrease the occurrence of ADRs and enhance the efficacy of anti-TB drugs. In this study, we used StellarPGx to call variants and star alleles in NAT1, NAT2, GSTM1, GSTT1, GSTP1, and CYP2E1, from 1079 high-depth African whole genomes. We present the distribution of common, rare, and potential novel star alleles across various Sub-Saharan African (SSA) populations, in comparison with other global populations. NAT1*10 (53.6%), GSTT1*0 (65%), GSTM1*0 (48%), and NAT2*5 (17.5%) were among the predominant functionally relevant star alleles. Additionally, we predicted varying phenotype distributions for NAT1 and NAT2 (acetylation) and the glutathione-S-transferase (GST) enzymes (detoxification activity) between SSA and other global populations. Forty-seven potentially novel haplotypes were identified computationally across the genes. This study provides insight into the distribution of key variants and star alleles potentially relevant to anti-TB drug metabolism and other drugs prescribed across various African populations. The high number of potentially novel star alleles exemplifies the need for pharmacogenomics studies in the African context. Overall, our study provides a foundation for functional pharmacogenetic studies and potential implementation of pharmacogenetic testing in Africa to reduce the risk of ADRs related to treatment of TB and other diseases.
Collapse
Affiliation(s)
- Thandeka V.B. Malinga
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
- Division of Human Genetics, National Health Laboratory Service, and School of Pathology, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Houcemeddine Othman
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
- Laboratory of Cytogenetics, Molecular Genetics and Reproductive Biology (LR03SP02)Farhat Hached University HospitalSousseTunisia
| | - Maria Paximadis
- School of Molecular and Cell Biology, Faculty of ScienceUniversity of the WitwatersrandJohannesburgSouth Africa
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services and Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Caroline T. Tiemessen
- Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services and Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Michèle Ramsay
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Scott Hazelhurst
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
- School of Electrical and Information EngineeringUniversity of the WitwatersrandJohannesburgSouth Africa
| | - David Twesigomwe
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
14
|
Martins Rodrigues F, Terekhanova NV, Imbach KJ, Clauser KR, Esai Selvan M, Mendizabal I, Geffen Y, Akiyama Y, Maynard M, Yaron TM, Li Y, Cao S, Storrs EP, Gonda OS, Gaite-Reguero A, Govindan A, Kawaler EA, Wyczalkowski MA, Klein RJ, Turhan B, Krug K, Mani DR, Leprevost FDV, Nesvizhskii AI, Carr SA, Fenyö D, Gillette MA, Colaprico A, Iavarone A, Robles AI, Huang KL, Kumar-Sinha C, Aguet F, Lazar AJ, Cantley LC, Marigorta UM, Gümüş ZH, Bailey MH, Getz G, Porta-Pardo E, Ding L. Precision proteogenomics reveals pan-cancer impact of germline variants. Cell 2025; 188:2312-2335.e26. [PMID: 40233739 DOI: 10.1016/j.cell.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/29/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
We investigate the impact of germline variants on cancer patients' proteomes, encompassing 1,064 individuals across 10 cancer types. We introduced an approach, "precision peptidomics," mapping 337,469 coding germline variants onto peptides from patients' mass spectrometry data, revealing their potential impact on post-translational modifications, protein stability, allele-specific expression, and protein structure by leveraging the relevant protein databases. We identified rare pathogenic and common germline variants in cancer genes potentially affecting proteomic features, including variants altering protein abundance and structure and variants in kinases (ERBB2 and MAP2K2) impacting phosphorylation. Precision peptidome analysis predicted destabilizing events in signal-regulatory protein alpha (SIRPA) and glial fibrillary acid protein (GFAP), relevant to immunomodulation and glioblastoma diagnostics, respectively. Genome-wide association studies identified quantitative trait loci for gene expression and protein levels, spanning millions of SNPs and thousands of proteins. Polygenic risk scores correlated with distal effects from risk variants. Our findings emphasize the contribution of germline genetics to cancer heterogeneity and high-throughput precision peptidomics.
Collapse
Affiliation(s)
- Fernanda Martins Rodrigues
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Nadezhda V Terekhanova
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kathleen J Imbach
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isabel Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Translational Prostate Cancer Research Lab, CIC bioGUNE-Basurto, Biocruces Bizkaia Health Research Institute, Derio, Spain
| | - Yifat Geffen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Yo Akiyama
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Tomer M Yaron
- Meyer Cancer Center, Department of Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Erik P Storrs
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Olivia S Gonda
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA
| | - Adrian Gaite-Reguero
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Akshay Govindan
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Emily A Kawaler
- Applied Bioinformatics Laboratories, New York University Langone Health, New York City, NY, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Berk Turhan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karsten Krug
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - D R Mani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Antonio Colaprico
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Antonio Iavarone
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD, USA
| | - Kuan-Lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Alexander J Lazar
- Departments of Pathology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Urko M Marigorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Matthew H Bailey
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA.
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Eduard Porta-Pardo
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Barcelona Supercomputing Center (BSC), Barcelona, Spain.
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, Saint Louis, MO, USA.
| |
Collapse
|
15
|
Nagasaki M, Hirayasu K, Khor SS, Otokozawa R, Sekiya Y, Kawai Y, Tokunaga K. JoGo-LILR caller: Unveiling and navigating the complex diversity of LILRB3-LILRA6 copy number haplotype structures with whole-genome sequencing. Hum Immunol 2025; 86:111272. [PMID: 40054016 DOI: 10.1016/j.humimm.2025.111272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 03/09/2025]
Abstract
Leukocyte immunoglobulin-like receptors (LILRs), encoded on human chromosome 19q13.4, comprise a set of 11 immunoglobulin superfamily receptors known for their genetic heterogeneity. Notably, LILRB3 and LILRA6 within this cluster exhibit pronounced sequence homology in immunoglobulin-like domains involved in ligand binding and variable copy number (CN) states. However, understanding their precise role remains challenging. To address this difficulty, we developed an algorithm and tool named JoGo-LILR Caller, which jointly calls CNs of LILRB3 and LILRA6 from a population-scale whole-genome short-read sequencing dataset. This tool was applied to 2,504 international HapMap samples and yielded a global CN profile. The 100 % concordance rate corroborated this profile with the CN data obtained from 40 samples of pangenome reference assemblies provided by the Human Pangenome Reference Consortium (HPRC). The frequencies of LILRB3-LILRA6 CN haplotype structures were also estimated for five continental groups with a global CN profile. The established allele frequency profile allowed our tool to estimate LILRB3-LILRA6 CN haplotype combinations. JoGo-LILR-trio enhanced the prediction reliability for haplotype pairs within trio datasets, with trio analysis on 40 child samples demonstrating a 100 % concordance between the predicted pair of haploid CN types and the diploid reference assemblies. Its utility will extend to facilitating software advancements for imputing LILRB3-LILRA6 CN types from SNP array genotyping data, enabling subsequent association analyses that link these CN types to diverse phenotypic traits and diseases, e.g., inflammatory bowel diseases and Takayasu arteritis.
Collapse
Affiliation(s)
- Masao Nagasaki
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan; Center for Genomic Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Kouyuki Hirayasu
- Advanced Preventive Medical Sciences Research Center, Kanazawa University, Kanazawa, Japan; Department of Evolutionary Immunology, Graduate School of Advanced Preventive Medical Sciences, Kanazawa University, Kanazawa, Japan; Department of Immunology, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan; Department of Immunology, School of Medical and Pharmaceutical Sciences, Kanazawa University, Kanazawa, Japan
| | - Seik-Soon Khor
- Genome Medical Science Project, National Center for Global Health and Medicine, Tokyo, Japan; Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 637551 Singapore, Singapore
| | - Ryoko Otokozawa
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yayoi Sekiya
- Division of Biomedical Information Analysis, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yosuke Kawai
- Genome Medical Science Project, National Center for Global Health and Medicine, Tokyo, Japan
| | - Katsushi Tokunaga
- Genome Medical Science Project, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
16
|
Lanting P, Warmerdam R, Slager J, Brugge H, Ochi T, Benjamins M, Lopera-Maya E, Jankipersadsing S, Gelderloos-Arends J, Teuben D, Hendriksen D, Charbon B, Johansson L, Munnink TO, de Boer-Veger N, Wilffert B, Swertz M, Touw D, Deelen P, Knoers N, Dekens J, Franke L. Low-cost generation of clinical-grade, layperson-friendly pharmacogenetic passports using oligonucleotide arrays. Am J Hum Genet 2025; 112:1015-1028. [PMID: 40174590 DOI: 10.1016/j.ajhg.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 04/04/2025] Open
Abstract
Pharmacogenomic (PGx) information is essential for precision medicine, enabling drug prescriptions to be personalized according to an individual's genetic background. Almost all individuals will carry a genetic marker that affects their drug response, so the ideal drug prescription for these individuals will differ from the population-level guidelines. Currently, PGx information is often not available at first prescription, reducing its effectiveness. In the Netherlands, pharmacogenetic information is most often obtained using dedicated single-gene assays, making it expensive and time consuming to generate complete multi-gene PGx profiles. We therefore hypothesized that we could also use genome-wide oligonucleotide genotyping arrays to generate comprehensive PGx information (PGx passports), thereby decreasing the cost and time required for PGx testing and lowering the barrier to generating PGx information prior to first prescription. Taking advantage of existing genetic data generated in two biobanks, we developed and validated Asterix, a low-cost, clinical-grade PGx passport pipeline for 12 PGx genes. In these biobanks, we performed and clinically validated genetic variant calling and statistical phasing and imputation. In addition, we developed and validated a CYP2D6 copy-number-variant-calling tool, forgoing the need to use separate PCR-based copy-number detection. Ultimately, we returned 1,227 PGx passports to biobank participants via a layperson-friendly app, improving knowledge of PGx among citizens. Our study demonstrates the feasibility of a low-cost, clinical-grade PGx passport pipeline that could be readily implemented in clinical settings to enhance personalized healthcare, ensuring that patients receive the most effective and safe drug therapy based on their unique genetic makeup.
Collapse
Affiliation(s)
- Pauline Lanting
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Robert Warmerdam
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Jelle Slager
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Harm Brugge
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Taichi Ochi
- Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, Epidemiology & Economics, University of Groningen, Groningen, the Netherlands
| | - Marloes Benjamins
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Esteban Lopera-Maya
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Soesma Jankipersadsing
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jody Gelderloos-Arends
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Daphne Teuben
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Dennis Hendriksen
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Genomics Coordination Center, Groningen, the Netherlands
| | - Bart Charbon
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Genomics Coordination Center, Groningen, the Netherlands
| | - Lennart Johansson
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Genomics Coordination Center, Groningen, the Netherlands
| | - Thijs Oude Munnink
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Bob Wilffert
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Groningen Research Institute of Pharmacy, Unit of PharmacoTherapy, Epidemiology & Economics, University of Groningen, Groningen, the Netherlands
| | - Morris Swertz
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Genomics Coordination Center, Groningen, the Netherlands
| | - Daan Touw
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Patrick Deelen
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Nine Knoers
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Jackie Dekens
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Center for Development and Innovation, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - Lude Franke
- Department of Genetics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| |
Collapse
|
17
|
Tafazoli A, Hemmati M, Rafigh M, Alimardani M, Khaghani F, Korostyński M, Karnes JH. Leveraging long-read sequencing technologies for pharmacogenomic testing: applications, analytical strategies, challenges, and future perspectives. Front Genet 2025; 16:1435416. [PMID: 40370700 PMCID: PMC12075302 DOI: 10.3389/fgene.2025.1435416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 04/07/2025] [Indexed: 05/16/2025] Open
Abstract
Long-read sequencing (LRS) was introduced as the third generation of next-generation sequencing technologies with a high accuracy rate in genomic variant identification for some of its platforms. Due to the structural complexity of many pharmacogenes, the presence of rare variants, and the limitations of genotyping and short-read sequencing approaches in detecting pharmacovariants, LRS methods are likely to become increasingly utilized in the near future. In this review, we aim to provide a comprehensive discussion of current and future applications of long-read genotyping methods by introducing the opportunities and advantages as well as the challenges and disadvantages of state-of-the-art LRS platforms for the implementation of pharmacogenomic tests in clinical and research settings. New approaches to data processing, as well as the challenges and pitfalls of performing such tests in daily practice, will be explored in detail. We provide references to resources for those who are interested or intend to employ LRS in pharmacogenomics screening, both in clinical and research settings.
Collapse
Affiliation(s)
- Alireza Tafazoli
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Mahboobeh Hemmati
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahboobeh Rafigh
- Medical Genetics Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maliheh Alimardani
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Faeze Khaghani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Michał Korostyński
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Jason H. Karnes
- Department of Pharmacy Practice and Science, University of Arizona R. Ken Coit College of Pharmacy, Tucson, AZ, United States
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
18
|
Pinotti T, Adler MA, Mermejo R, Bitz-Thorsen J, McColl H, Scorrano G, Feizabadifarahani M, Gandy D, Boulanger M, Gaunitz C, Stenderup J, Ramsøe A, Korneliussen T, Demeter F, Santos FR, Vinner L, Sikora M, Meltzer DJ, Moreno-Mayar JV, Quanchello C, Willerslev E. Picuris Pueblo oral history and genomics reveal continuity in US Southwest. Nature 2025:10.1038/s41586-025-08791-9. [PMID: 40307544 DOI: 10.1038/s41586-025-08791-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 02/14/2025] [Indexed: 05/02/2025]
Abstract
Indigenous groups often encounter significant challenges when asserting ancestral claims and cultural affiliations based on oral histories, particularly in the USA where such narratives have historically been undervalued. Although ancient DNA offers a tool to complement traditional knowledge and address gaps in oral history, longstanding disregard for Indigenous sovereignty and beliefs has understandably led many Indigenous communities to distrust DNA studies1-4. Earlier research often focused on repatriation claims5-7, whereas more recent work has increasingly moved towards enhancing Tribal histories8,9. Here we present a collaborative study initiated by a federally recognized Native American tribe, the sovereign nation of Picuris Pueblo in the Northern Rio Grande region of New Mexico, USA, to address gaps in traditional knowledge and further their understanding of their population history and ancestry. We generated genomes from 16 ancient Picuris individuals and 13 present-day members of Picuris Pueblo, providing genomic data spanning the last millennium. We show genetic continuity between ancient and present-day Picuris, and more broadly with Ancestral Puebloans from Pueblo Bonito in Chaco Canyon10, 275 km to the west. This suggests a firm spatiotemporal link among these Puebloan populations of the North American Southwest. Furthermore, we see no evidence of population decline before European arrival11-13, and no Athabascan ancestry in individuals predating 1500 CE, challenging earlier migration hypotheses14-16. This work prioritizes Indigenous control of genetic data and brings together oral tradition, archaeology, ethnography and genetics.
Collapse
Affiliation(s)
- Thomaz Pinotti
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Laboratório de Biodiversidade e Evolução Molecular (LBEM), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Michael A Adler
- Department of Anthropology, Southern Methodist University, Dallas, TX, USA
| | | | - Julie Bitz-Thorsen
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Hugh McColl
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Gabriele Scorrano
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Center for Molecular Anthropology for the Study of Ancient DNA, Department of Biology, University of Rome 'Tor Vergata', Rome, Italy
| | - Motahareh Feizabadifarahani
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Department of Archaeology and Classical Studies, Stockholm University, Stockholm, Sweden
| | - Devlin Gandy
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Department of Archaeology, University of Cambridge, Cambridge, UK
| | - Matthew Boulanger
- Department of Anthropology, Southern Methodist University, Dallas, TX, USA
| | - Charleen Gaunitz
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Stenderup
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Abigail Ramsøe
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Thorfinn Korneliussen
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Fabrice Demeter
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Eco-anthropologie (EA), Departement ABBA, Muséum national d'Histoire naturelle, CNRS, Université Paris Cité, Musée de l'Homme, Paris, France
| | - Fabrício R Santos
- Laboratório de Biodiversidade e Evolução Molecular (LBEM), Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lasse Vinner
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - Martin Sikora
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | - David J Meltzer
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
- Department of Anthropology, Southern Methodist University, Dallas, TX, USA
| | - J Víctor Moreno-Mayar
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark
| | | | - Eske Willerslev
- Lundbeck Foundation GeoGenetics Centre, GLOBE Institute, University of Copenhagen, Copenhagen, Denmark.
- Department of Genetics, University of Cambridge, Cambridge, UK.
- MARUM Center for Marine Environmental Sciences, University of Bremen, Bremen, Germany.
| |
Collapse
|
19
|
Xie L, Peng YQ, Shen X. Identifying therapeutic target genes for diabetic retinopathy using systematic druggable genome-wide Mendelian randomization. Diabetol Metab Syndr 2025; 17:145. [PMID: 40301928 PMCID: PMC12039192 DOI: 10.1186/s13098-025-01710-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
INTRODUCTION The treatment and prevention of diabetic retinopathy (DR) remain significant challenges. Mendelian randomization (MR) has been widely used to explore novel therapeutic targets. In this study, we conducted a systematic druggable genome-wide MR analysis to explore potential therapeutic targets for DR. METHODS We obtained data on druggable genes and screened for genes within blood expression quantitative trait loci (eQTL), which were then subjected to MR analysis and colocalization analysis with DR genome-wide association studies data to identify genes strongly associated with DR. Additionally, Gene Ontology (GO) analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, protein-protein interaction (PPI) network construction, drug candidate prediction, and molecular docking were performed to provide valuable insights for the development of more effective and targeted therapeutic drugs. RESULTS MR analysis of blood eQTLs revealed 30 significant DR-associated druggable genes, with PRKAB1 (OR = 0.935, 95% CI: 0.892 to 0.980) and CNR1 (OR = 0.814, 95% CI: 0.696 to 0.951) being protective genes, whereas CACNA1E (OR = 1.282, 95% CI: 1.050 to 1.565), NME1 (OR = 1.198, 95% CI: 1.028 to 1.397), and CHRNA2 (OR = 1.192, 95% CI: 1.025 to 1.386) were associated with increased risk. KEGG analysis highlighted significant pathways, including adrenergic signaling in cardiomyocytes (hsa04261), the oxytocin signaling pathway (hsa04921), and arrhythmogenic right ventricular cardiomyopathy (hsa05412). PPI network analysis identified two key modules: one comprising BIN1, CDH2, ACTN1, EPAS1, CEBPA, and CTSD nodes, and the other consisting of CACNG6, CACNA1E, CACNA2D3, and RASGRP3 nodes. Drug candidate prediction suggested ethanol and isoflupredone as potential therapeutic interventions, and molecular docking revealed C5's strong protein binding affinity. CONCLUSIONS This study utilized MR and colocalization analysis to identify potential drug targets for DR. The findings provide promising leads for the treatments of DR, potentially reducing drug development costs.
Collapse
Affiliation(s)
- Long Xie
- Department of Orthopedics, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha), Hunan Normal University, Changsha, Hunan Province, 410006, China.
| | - Yu Qin Peng
- Department of Ophthalmology, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha), Hunan Normal University, Changsha, Hunan Province, 410006, China
| | - Xiang Shen
- Department of Orthopedics, The Fourth Hospital of Changsha (Integrated Traditional Chinese and Western Medicine Hospital of Changsha), Hunan Normal University, Changsha, Hunan Province, 410006, China
| |
Collapse
|
20
|
Ding F, Pang Z, Ji X, Jiang Y, Wang Q, Bing Z. Identification of Risk Loci for Radiotherapy-Induced Tinnitus and Hearing Loss Through Integrated Genomic Analysis. Int J Mol Sci 2025; 26:4132. [PMID: 40362371 PMCID: PMC12071707 DOI: 10.3390/ijms26094132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Radiotherapy-induced hearing impairment significantly affects patients' quality of life, yet its genetic basis remains poorly understood. This study seeks to identify genetic variants associated with radiotherapy-induced tinnitus and hearing loss and explore their functional implications. A genome-wide association study (GWAS) was conducted to identify single-nucleotide polymorphisms (SNPs) associated with radiotherapy-induced tinnitus and hearing loss. Protein-protein interaction networks and functional enrichment analyses were performed to explore underlying biological pathways. A phenome-wide association study (PheWAS) analysis across five databases examined associations between identified SNPs and various phenotypes. The GWAS identified 97 SNPs significantly associated with radiotherapy-induced tinnitus and 76 SNPs with hearing loss. Tinnitus-associated variants were enriched in pathways involving Wnt signaling and telomerase RNA regulation, while hearing-loss-associated variants were linked to calcium-dependent cell adhesion and neurotransmitter receptor regulation. The PheWAS analysis revealed significant associations between these hearing-impairment-related SNPs and metabolic phenotypes, particularly BMI and metabolic disorders. A chromosomal distribution analysis showed concentrated significant SNPs on chromosomes 1, 2, 5, and 10. This study identified distinct genetic architectures underlying radiotherapy-induced tinnitus and hearing loss, revealing different molecular pathways involved in their pathogenesis. The unexpected association with metabolic phenotypes suggests potential interactions between metabolic status and susceptibility to radiotherapy-induced hearing complications. These findings provide insights for developing genetic screening tools and targeted interventions to prevent or mitigate radiotherapy-related hearing damage.
Collapse
Affiliation(s)
- Fan Ding
- Teaching and Experimental Training Center, Gansu University of Chinese Medicine, Lanzhou 730000, China; (Z.P.); (X.J.); (Y.J.); (Q.W.)
| | - Zehao Pang
- Teaching and Experimental Training Center, Gansu University of Chinese Medicine, Lanzhou 730000, China; (Z.P.); (X.J.); (Y.J.); (Q.W.)
| | - Xiujia Ji
- Teaching and Experimental Training Center, Gansu University of Chinese Medicine, Lanzhou 730000, China; (Z.P.); (X.J.); (Y.J.); (Q.W.)
| | - Yuanfang Jiang
- Teaching and Experimental Training Center, Gansu University of Chinese Medicine, Lanzhou 730000, China; (Z.P.); (X.J.); (Y.J.); (Q.W.)
| | - Qiulan Wang
- Teaching and Experimental Training Center, Gansu University of Chinese Medicine, Lanzhou 730000, China; (Z.P.); (X.J.); (Y.J.); (Q.W.)
| | - Zhitong Bing
- Advanced Nuclear Physics Laboratory, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| |
Collapse
|
21
|
Toki R, Fushiki S, Kojima S, Sutoh Y, Otsuka-Yamasaki Y, Harada S, Iida M, Hirata A, Miyagawa N, Matsumoto M, Edagawa S, Miyake A, Kuwabara K, Hirayama A, Sugimoto M, Sato A, Amano K, Soga T, Tomita M, Arakawa K, Kinoshita K, Sakurai-Yageta M, Tamiya G, Ohmomo H, Shimizu A, Okamura T, Takebayashi T. Genome-wide association study of plasma amino acids and Mendelian randomization for cardiometabolic traits. Sci Rep 2025; 15:14569. [PMID: 40281240 PMCID: PMC12032298 DOI: 10.1038/s41598-025-98992-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
Plasma amino acids (AAs) have emerged as promising biomarkers for metabolic disorders, yet their causality remains unclear. We aimed to investigate the genetic determinants of AA levels in a cohort of 10,333 individuals and their causal effects on cardiometabolic traits using Mendelian randomization (MR). Plasma levels of 20 AAs were quantified using capillary electrophoresis mass spectrometry. Genome-wide association studies were conducted using BOLT-LMM and heritability estimation via LDSC analysis. Causal effects of AAs on 11 cardiometabolic traits were examined using two-sample MR analyses. We identified 85 locus-metabolite associations across 43 genes for 18 AAs, including 44 novel loci linked to metabolic genes. Heritability for AAs was estimated at 16%. MR analysis demonstrated cystine to positively associate with systolic blood pressure (SBP) (β = 0.056, SE = 0.010), while serine indicated protective effects on SBP (β = - 0.040, SE = 0.011), diastolic BP (β = - 0.044, SE = 0.010), and coronary artery disease (odds ratio 0.888, SE = 0.028). We identified potentially novel genetic loci associated with AA levels and demonstrated robust causal associations between several AAs and cardiometabolic traits. These findings reinforce the importance of AAs as potential biomarkers and therapeutic targets in cardiometabolic health.
Collapse
Affiliation(s)
- Ryota Toki
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Sotaro Fushiki
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
- Konica Minolta, Inc, Chiyoda-ku, Tokyo, Japan
| | - Shun Kojima
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
- Konica Minolta, Inc, Chiyoda-ku, Tokyo, Japan
| | - Yoichi Sutoh
- Division of Bioinformation Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Shiwa-gun, Iwate, Japan
- Division of Bioinformation Analysis, Institute for Biomedical Sciences, Iwate Medical University, Shiwa-gun, Iwate, Japan
| | - Yayoi Otsuka-Yamasaki
- Division of Bioinformation Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Shiwa-gun, Iwate, Japan
- Division of Bioinformation Analysis, Institute for Biomedical Sciences, Iwate Medical University, Shiwa-gun, Iwate, Japan
| | - Sei Harada
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Miho Iida
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Aya Hirata
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Naoko Miyagawa
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Minako Matsumoto
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Shun Edagawa
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Atsuko Miyake
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Kazuyo Kuwabara
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Tsuruoka City, Yamagata, Japan
| | - Masahiro Sugimoto
- Institute for Advanced Biosciences, Keio University, Tsuruoka City, Yamagata, Japan
| | - Asako Sato
- Institute for Advanced Biosciences, Keio University, Tsuruoka City, Yamagata, Japan
| | - Kaori Amano
- Institute for Advanced Biosciences, Keio University, Tsuruoka City, Yamagata, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Tsuruoka City, Yamagata, Japan
| | - Masaru Tomita
- Institute for Advanced Biosciences, Keio University, Tsuruoka City, Yamagata, Japan
| | - Kazuharu Arakawa
- Institute for Advanced Biosciences, Keio University, Tsuruoka City, Yamagata, Japan
| | - Kengo Kinoshita
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Information Sciences, Tohoku University, Sendai, Miyagi, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi, Japan
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi, Japan
| | - Mika Sakurai-Yageta
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai, Miyagi, Japan
| | - Gen Tamiya
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
- Graduate School of Medicine, Tohoku University, Sendai, Miyagi, Japan
- Center for Advanced Intelligence Project, RIKEN, Wako, Saitama, Japan
| | - Hideki Ohmomo
- Division of Bioinformation Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Shiwa-gun, Iwate, Japan
- Division of Bioinformation Analysis, Institute for Biomedical Sciences, Iwate Medical University, Shiwa-gun, Iwate, Japan
| | - Atsushi Shimizu
- Division of Bioinformation Analysis, Iwate Tohoku Medical Megabank Organization, Iwate Medical University, Shiwa-gun, Iwate, Japan
- Division of Bioinformation Analysis, Institute for Biomedical Sciences, Iwate Medical University, Shiwa-gun, Iwate, Japan
| | - Tomonori Okamura
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan
| | - Toru Takebayashi
- Department of Preventive Medicine and Public Health, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, Japan.
| |
Collapse
|
22
|
Garcia-Calleja J, Biagini SA, de Cid R, Calafell F, Bosch E. Inferring past demography and genetic adaptation in Spain using the GCAT cohort. Sci Rep 2025; 15:14225. [PMID: 40274920 PMCID: PMC12022144 DOI: 10.1038/s41598-025-98272-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Located in the southwestern corner of Europe, the Iberian Peninsula is separated from the rest of the continent by the Pyrenees Mountains and from Africa by the Strait of Gibraltar. This geographical position may have conditioned distinct selective pressures compared to the rest of Europe and influenced differential patterns of gene flow. In this work, we analyse 704 whole-genome sequences from the GCAT reference panel to quantify gene flow into Spain from various historical sources and identify the top signatures of positive (adaptive) selection. While we found no clear evidence of a 16th-century admixture event putatively related to the French diaspora during the Wars of Religion, we detected signals of North African admixture matching the Muslim period and the subsequent Christian Reconquista. Notably, besides finding that well-known candidate genes previously described in Eurasians also seem to be adaptive in Spain, we discovered novel top candidates for positive selection putatively associated with immunity and diet (UBL7, SMYD1, VAC14 and FDFT1). Finally, local ancestry deviation analysis revealed that the MHCIII genomic region underwent post-admixture selection following the post-Neolithic admixture with Steppe ancestry.
Collapse
Affiliation(s)
- Jorge Garcia-Calleja
- Institute of Evolutionary Biology (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
| | - Simone A Biagini
- Institute of Evolutionary Biology (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain
- Department of Archaeology and Museology, Masaryk University, Brno, Czech Republic
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Rafael de Cid
- Genomes for Life-GCAT lab, CORE Program, Germans Trias i Pujol Research Institute (IGTP), 08916, Badalona, Spain
- Grup de REcerca en Impacte de les Malalties Cròniques i les seves Trajectòries (GRIMTra), Germans Trias I Pujol Research Institute (IGTP), 08916, Badalona, Spain
| | - Francesc Calafell
- Institute of Evolutionary Biology (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain.
| | - Elena Bosch
- Institute of Evolutionary Biology (UPF-CSIC), Department of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain.
| |
Collapse
|
23
|
Omae Y, Khor SS, Shimada M, Kawai Y, Yamaguchi T, Yagi M, Ebisawa M, Takeuchi JS, Mizoue T, Sugiura W, Tokunaga K. Genome-wide association study of common side effects following COVID-19 booster vaccination in a cohort of corporate employees in Japan. Sci Rep 2025; 15:12728. [PMID: 40222985 PMCID: PMC11994816 DOI: 10.1038/s41598-025-90787-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/17/2025] [Indexed: 04/15/2025] Open
Abstract
Individual differences have been observed in side effects after vaccination for COVID-19, and host genetic factors have been suggested as a contributing factor. Here, we conducted a genome-wide association study (GWAS) involving 2,554 Japanese corporate employees who received a third booster dose of BNT162b2/Pfizer or mRNA-1273/Moderna vaccine. Although no genome-wide significant association was found for the presence of adverse symptoms, the GWAS for severity revealed six associated loci. The most significant association was observed between the severity of swelling of lymph nodes and chromosome 2q12 locus, including the IL1RL1, IL18R1, and IL18RAP genes (lead variant: rs76152249; P = 1.46 × 10-9). Pathway analysis suggested associations between immune pathways related to the MHC locus, including HLA genes, and the occurrence and severity of fever, and the NF-κB binding pathway and those of itching at the injection site. In addition, a meta-analysis of previous GWAS studies for the primary first or second dose of COVID-19 vaccine revealed 818 variants from 72 loci that demonstrated genome-wide significant associations with any of 12 symptoms, and pathway analysis identified immune pathways related to the MHC locus, suggesting shared genetic risks among primary and booster vaccinations. These results may help control side effects following COVID-19 vaccination.
Collapse
Affiliation(s)
- Yosuke Omae
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Seik-Soon Khor
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, Singapore, Singapore
| | - Mihoko Shimada
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yosuke Kawai
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Taihei Yamaguchi
- Life Science Business Office, Corporate Technology Planning Division, Toshiba Corporation, Tokyo, Japan
| | - Maiko Yagi
- Life Science Business Office, Corporate Technology Planning Division, Toshiba Corporation, Tokyo, Japan
| | - Masashi Ebisawa
- Life Science Business Office, Corporate Technology Planning Division, Toshiba Corporation, Tokyo, Japan
| | - Junko S Takeuchi
- Department of Academic-Industrial Partnerships Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tetsuya Mizoue
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Wataru Sugiura
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Katsushi Tokunaga
- Genome Medical Science Project, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan.
| |
Collapse
|
24
|
Ma W, Chaisson M. Genotyping sequence-resolved copy number variation using pangenomes reveals paralog-specific global diversity and expression divergence of duplicated genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.11.607269. [PMID: 39149335 PMCID: PMC11326217 DOI: 10.1101/2024.08.11.607269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Copy number variant (CNV) genes are important in evolution and disease, yet sequence variation in CNV genes remains a blind spot in large-scale studies. We present ctyper, a method that leverages pangenomes to produce allele-specific copy numbers with locally phased variants from next-generation sequencing (NGS) reads. Benchmarking on 3,351 CNV genes, including HLA, SMN, and CYP2D6, and 212 challenging medically relevant (CMR) genes that are poorly mapped by NGS, ctyper captures 96.5% of phased variants with ≥99.1% correctness of copy number on CNV genes and 94.8% of phased variants on CMR genes. Applying alignment-free algorithms, ctyper requires 1.5 hours per genome on a single CPU. The results improve prediction of gene expression compared to known expression quantitative trait loci (eQTL) variants. Allele-specific expression quantified divergent expression on 7.94% of paralogs and tissue-specific biases on 4.68% of paralogs. We found reduced expression of SMN-2 due to SMN1 conversion, potentially affecting spinal muscular atrophy, and increased expression of translocated duplications of AMY2B. Overall, ctyper enables biobank-scale genotyping of CNV and CMR genes.
Collapse
|
25
|
Antonatos C, Mitsoudi D, Pontikas A, Akritidis A, Xiropotamos P, Georgakilas GK, Georgiou S, Tsiogka A, Gregoriou S, Grafanaki K, Vasilopoulos Y. Transcriptome-wide analyses delineate the genetic architecture of expression variation in atopic dermatitis. HGG ADVANCES 2025; 6:100422. [PMID: 40017037 PMCID: PMC11937661 DOI: 10.1016/j.xhgg.2025.100422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/01/2025] Open
Abstract
Genome-wide association studies (GWASs) for atopic dermatitis (AD) have uncovered 81 risk loci in European participants; however, translating these findings into functional and therapeutic insights remains challenging. We conducted a transcriptome-wide association study (TWAS) in AD leveraging cis-eQTL data from sun exposed (n = 517), non-sun exposed skin (n = 602) and whole blood (n = 670) tissues and the latest GWAS of AD in Europeans (n = 864982). We implemented the OTTERS pipeline that combines polygenic risk score (PRS) techniques accommodating diverse assumptions in the architecture of gene regulation. We also used differential expression meta-analysis and co-expression networks (n = 186) to characterize the transcriptomic landscape of AD. We identified 176 gene-tissue associations covering 126 unique genes (53 previously unreported). Most TWAS risk genes were identified by adaptive PRS frameworks, with non-significant differences compared with clumping and thresholding approaches. TWAS risk genes were enriched in allergic reactions (e.g., AQP7, AFF4), skin barrier integrity (e.g., ACER3), and inflammatory pathways (e.g., TAPBPL). By integrating co-expression networks of lesional AD skin, we identified 16 hub genes previously identified as TWAS risk genes (six previously unreported) that orchestrate inflammatory responses (e.g., HSPA4) and keratinization (e.g., LCE3E, LCE3D), serving as potential drug targets through drug-gene interactions. Consistent associations between all analyses were reported for FOSL1 and RORC. Collectively, our findings provide additional risk genes for AD with potential implications in therapeutic approaches.
Collapse
Affiliation(s)
- Charalabos Antonatos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Dimitra Mitsoudi
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Alexandros Pontikas
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Adam Akritidis
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece
| | - Panagiotis Xiropotamos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece; Information Management Systems Institute, ATHENA Research Center, 15125 Marousi, Greece
| | - Georgios K Georgakilas
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece; Information Management Systems Institute, ATHENA Research Center, 15125 Marousi, Greece
| | - Sophia Georgiou
- Department of Dermatology-Venereology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Aikaterini Tsiogka
- Department of Dermatology-Venereology, Faculty of Medicine, Andreas Sygros Hospital, National and Kapodistrian University of Athens, 16121 Athens, Greece
| | - Stamatis Gregoriou
- Department of Dermatology-Venereology, Faculty of Medicine, Andreas Sygros Hospital, National and Kapodistrian University of Athens, 16121 Athens, Greece
| | - Katerina Grafanaki
- Department of Dermatology-Venereology, School of Medicine, University of Patras, 26504 Patras, Greece; Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Yiannis Vasilopoulos
- Laboratory of Genetics, Section of Genetics, Cell Biology and Development, Department of Biology, University of Patras, 26504 Patras, Greece.
| |
Collapse
|
26
|
Xu J, Liu D, Hassan A, Genovese G, Cote AC, Fennessy B, Cheng E, Charney AW, Knowles JA, Ayub M, Peterson RE, Bigdeli TB, Huckins LM. Evaluation of imputation performance of multiple reference panels in a Pakistani population. HGG ADVANCES 2025; 6:100395. [PMID: 39696820 PMCID: PMC11759560 DOI: 10.1016/j.xhgg.2024.100395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/16/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
Genotype imputation is crucial for genome-wide association studies (GWASs), but reference panels and existing benchmarking studies prioritize European individuals. Consequently, it is unclear which publicly available reference panel should be used for Pakistani individuals, and whether ancestry composition or sample size of the panel matters more for imputation accuracy. Our study compared different reference panels to impute genotype data in 1,814 Pakistani individuals, finding the best performance balancing accuracy and coverage with meta-imputation with TOPMed and the expanded 1000 Genomes (ex1KG) reference. Imputation accuracy of ex1KG outperformed TOPMed for common variants despite its 30-fold smaller sample size, supporting efforts to create future panels with diverse populations.
Collapse
Affiliation(s)
- Jiayi Xu
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Dongjing Liu
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Arsalan Hassan
- University of Peshawar, Khyber Pakhtunkhwa, Peshawar 25120, Pakistan; Institute of Omics and Health Research, Lahore, Pakistan
| | - Giulio Genovese
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Alanna C Cote
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Brian Fennessy
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Esther Cheng
- Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | | | - James A Knowles
- The Human Genetics Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
| | | | - Roseann E Peterson
- Department of Psychiatry and Behavioral Sciences, Institute for Genomics in Health, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Tim B Bigdeli
- Department of Psychiatry and Behavioral Sciences, Institute for Genomics in Health, State University of New York Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Laura M Huckins
- Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
27
|
Carioscia SA, Biddanda A, Starostik MR, Tang X, Hoffmann ER, Demko ZP, McCoy RC. Common variation in meiosis genes shapes human recombination phenotypes and aneuploidy risk. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.02.25325097. [PMID: 40321295 PMCID: PMC12047964 DOI: 10.1101/2025.04.02.25325097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
The leading cause of human pregnancy loss is aneuploidy, often tracing to errors in chromosome segregation during female meiosis. While abnormal crossover recombination is known to confer risk for aneuploidy, limited data have hindered understanding of the potential shared genetic basis of these key molecular phenotypes. To address this gap, we performed retrospective analysis of preimplantation genetic testing data from 139,416 in vitro fertilized embryos from 22,850 sets of biological parents. By tracing transmission of haplotypes, we identified 3,656,198 crossovers, as well as 92,485 aneuploid chromosomes. Counts of crossovers were lower in aneuploid versus euploid embryos, consistent with their role in chromosome pairing and segregation. Our analyses further revealed that a common haplotype spanning the meiotic cohesin SMC1B is significantly associated with both crossover count and maternal meiotic aneuploidy, with evidence supporting a non-coding cis-regulatory mechanism. Transcriptome- and phenome-wide association tests also implicated variation in the synaptonemal complex component C14orf39 and crossover-regulating ubiquitin ligases CCNB1IP1 and RNF212 in meiotic aneuploidy risk. More broadly, recombination and aneuploidy possess a partially shared genetic basis that also overlaps with reproductive aging traits. Our findings highlight the dual role of recombination in generating genetic diversity, while ensuring meiotic fidelity.
Collapse
Affiliation(s)
| | - Arjun Biddanda
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | | | - Xiaona Tang
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Eva R. Hoffmann
- DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Rajiv C. McCoy
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
28
|
Luan B, Yang Y, Yang Q, Li Z, Xu Z, Chen Y, Wang M, Chen W, Ge F. Gut microbiota, blood metabolites, & pan-cancer: a bidirectional Mendelian randomization & mediation analysis. AMB Express 2025; 15:59. [PMID: 40175810 PMCID: PMC11965084 DOI: 10.1186/s13568-025-01866-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 03/14/2025] [Indexed: 04/04/2025] Open
Abstract
We propose using Mendelian randomization analysis on GWAS data and MetaboAnalyst to model gut microbiota, metabolic pathways, blood metabolites, and cancer risk. We examined 473 gut microbiota, 205 pathways, 1400 metabolites, and 8 cancers. Results were validated through bidirectional two-sample Mendelian Randomization (MR), heterogeneity tests, and pathway enrichment, leading to a mediation pathway model. We identified 129 gut microbiota, 57 pathways, and 463 metabolites linked to cancer, and 34 significant plasma pathways. 15 microbiota, 8 pathways, and 58 metabolites implicated in multiple cancers. Eight plasma metabolic pathways are involved in the development of multiple types of cancer. Through Multivariate Mendelian Randomization (MVMR) and mediation analysis, we found 9 mediation pathways, offering novel targets and research directions for cancer pathogenesis and treatment.
Collapse
Affiliation(s)
- Biqing Luan
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yang Yang
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Department of breast surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, Yunnan, China
| | - Qizhi Yang
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhiqiang Li
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Zhihui Xu
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yaqin Chen
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Meiting Wang
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenlin Chen
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Department of breast surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, Yunnan, China.
| | - Fei Ge
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
29
|
Paramita RI, Panigoro SS, Fadilah F, Wanandi SI, Sutandyo N. SNP-array profiling data from breast cancer patients and healthy women's blood DNA samples. Data Brief 2025; 59:111343. [PMID: 39990126 PMCID: PMC11847265 DOI: 10.1016/j.dib.2025.111343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/25/2025] Open
Abstract
Breast cancer is commonly acknowledged as the primary type of cancer on a global scale, exerting a substantial influence on death rates, particularly in developing countries. The aforementioned discovery provides evidence in favor of the concept that genetic factors may contribute to the onset of breast cancer. This paper presents the unprocessed idat data containing single nucleotide polymorphisms (SNPs) acquired from breast cancer patients and a control group comprising of healthy women. The DNA was obtained from stored blood samples that were collected from a total of 48 female patients diagnosed with breast cancer at Cipto Mangunkusumo National Hospital Jakarta and Dharmais National Cancer Center Hospital Jakarta. Additionally, 24 healthy women were included as control subjects. Subsequently, the DNA samples were subjected to hybridization onto Infinium Asian Screening Array (ASA)'s beadchips. The chip was then subjected to fluorescence intensity measurements using an iScan machine manufactured by Illumina. The data output is produced in the form of a .idat file for each sample. Subsequently, further quality control measures and population stratification analysis were conducted using PLINK (v1.9). After the conclusion of the quality control procedure, 72 individuals and a dataset consisting of 424,285 genetic variants were selected for further analysis. The idat raw data files have been added to the Gene Expression Omnibus (GEO) with accession number: GSE245794 (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE245794).
Collapse
Affiliation(s)
- Rafika Indah Paramita
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 4, Jakarta, 10430, Indonesia
- Department of Medical Chemistry, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 4, Jakarta, 10430, Indonesia
- Bioinformatics Core Facilities-IMERI, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 6, Jakarta, 10430, Indonesia
| | - Sonar Soni Panigoro
- Master's Programme in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 4, Jakarta, 10430, Indonesia
- Surgical Oncology Division, Department of Surgery, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 6, Jakarta, 10430, Indonesia
| | - Fadilah Fadilah
- Department of Medical Chemistry, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 4, Jakarta, 10430, Indonesia
- Bioinformatics Core Facilities-IMERI, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 6, Jakarta, 10430, Indonesia
- Master's Programme in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 4, Jakarta, 10430, Indonesia
| | - Septelia Inawati Wanandi
- Master's Programme in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 4, Jakarta, 10430, Indonesia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 4, Jakarta, 10430, Indonesia
- Molecular Biology and Proteomics Core Facilities-IMERI, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 6, Jakarta, 10430, Indonesia
| | - Noorwati Sutandyo
- Department of Hematology and Medical Oncology, Dharmais National Cancer Center Hospital, Jalan Letjen S. Parman, Jakarta, 11420, Indonesia
- Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Jalan Salemba Raya number 4, Jakarta, 10430, Indonesia
| |
Collapse
|
30
|
Wang M, Chen H, Luo L, Huang Y, Duan S, Yuan H, Tang R, Liu C, He G. Forensic investigative genetic genealogy: expanding pedigree tracing and genetic inquiry in the genomic era. J Genet Genomics 2025; 52:460-472. [PMID: 38969261 DOI: 10.1016/j.jgg.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/07/2024]
Abstract
Genetic genealogy provides crucial insights into the complex biological relationships within contemporary and ancient human populations by analyzing shared alleles and chromosomal segments that are identical by descent to understand kinship, migration patterns, and population dynamics. Within forensic science, forensic investigative genetic genealogy (FIGG) has gained prominence by leveraging next-generation sequencing technologies and population-specific genomic resources, opening useful investigative avenues. In this review, we synthesize current knowledge, underscore recent advancements, and discuss the growing role of FIGG in forensic genomics. FIGG has been pivotal in revitalizing dormant inquiries and offering genetic leads in numerous cold cases. Its effectiveness relies on the extensive single-nucleotide polymorphism profiles contributed by individuals from diverse populations to specialized genomic databases. Advances in computational genomics and the growth of human genomic databases have spurred a profound shift in the application of genetic genealogy across forensics, anthropology, and ancient DNA studies. As the field progresses, FIGG is evolving from a nascent practice into a more sophisticated and specialized discipline, shaping the future of forensic investigations.
Collapse
Affiliation(s)
- Mengge Wang
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan 610041, China; Center for Archaeological Science, Sichuan University, Chengdu, Sichuan 610041, China; Anti-Drug Technology Center of Guangdong Province, Guangzhou, Guangdong 510000, China.
| | - Hongyu Chen
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan 610041, China; Center for Archaeological Science, Sichuan University, Chengdu, Sichuan 610041, China; Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lintao Luo
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan 610041, China; Center for Archaeological Science, Sichuan University, Chengdu, Sichuan 610041, China; Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuguo Huang
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan 610041, China; Center for Archaeological Science, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuhan Duan
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan 610041, China
| | - Huijun Yuan
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan 610041, China; Center for Archaeological Science, Sichuan University, Chengdu, Sichuan 610041, China.
| | - Renkuan Tang
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Chao Liu
- Anti-Drug Technology Center of Guangdong Province, Guangzhou, Guangdong 510000, China.
| | - Guanglin He
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan 610041, China; Center for Archaeological Science, Sichuan University, Chengdu, Sichuan 610041, China; Anti-Drug Technology Center of Guangdong Province, Guangzhou, Guangdong 510000, China.
| |
Collapse
|
31
|
Zheng W, Ma W, Chen Z, Wang C, Sun T, Dong W, Zhang W, Zhang S, Tang Z, Li K, Zhao Y, Liu Y. DPImpute: A Genotype Imputation Framework for Ultra-Low Coverage Whole-Genome Sequencing and its Application in Genomic Selection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412482. [PMID: 40013759 PMCID: PMC12021046 DOI: 10.1002/advs.202412482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/05/2025] [Indexed: 02/28/2025]
Abstract
Whole-genome sequencing is pivotal for elucidating the complex relationships between genotype and phenotype. However, its widespread application is hindered by the high sequencing depth and large sample sizes required, especially for genomic selection (GS) reliant on precise phenotype prediction from high-density genotype data. To address this, DPImpute (Dual-Phase Impute) is developed, an two-step imputation pipeline enabling accurate whole-genome SNP genotyping under ultra-low coverage whole-genome sequencing (ulcWGS) depths, small testing sample sizes, and limited reference populations. DPImpute achieved 98.06% SNP imputation accuracy with minimal testing samples (≤10), reference samples (≤100), and an ultra-low sequencing depth of 0.3X, surpassing the accuracy of existing imputation methods. Moreover, this high accuracy is maintained across multi-ancestry human populations. Remarkably, DPImpute demonstrated accurate SNP imputation from low-coverage sequencing data from single blood cells and single blastocyst cells, highlighting its potential in embryo GS. To enhance the accessibility of DPImpute, a user-friendly web server (https://agdb.ecenr.com/DPImpute/home) is developed and a Docker container for seamless implementation. In summary, DPImpute can significantly expedite breeding programs through precise and cost-effective genotyping and serve as a valuable tool for diverse population genotyping, encompassing both human and animal studies.
Collapse
Affiliation(s)
- Weigang Zheng
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Wenlong Ma
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Zhilong Chen
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Chao Wang
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Tao Sun
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Wenjun Dong
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Wenjing Zhang
- State Key Laboratory of Swine and Poultry Breeding IndustryNational Engineering Research Center for Breeding Swine IndustryGuangdong Provincial Key Lab of Agro‐Animal Genomics and Molecular BreedingCollege of Animal ScienceSouth China Agricultural UniversityGuangzhou510642China
| | - Song Zhang
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Zhonglin Tang
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Kunpeng Institute of Modern Agriculture at FoshanChinese Academy of Agricultural SciencesFoshan528226China
| | - Kui Li
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
| | - Yunxiang Zhao
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and TechnologyGuangxi UniversityNanning530004China
| | - Yuwen Liu
- Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural AffairsCollege of Animal Science and TechnologyHuazhong Agricultural UniversityWuhan430070China
- Shenzhen BranchGuangdong Laboratory for Lingnan Modern AgricultureKey Laboratory of Livestock and Poultry Multi‐Omics of MARAAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Innovation Group of Pig Genome Design and BreedingResearch Centre for Animal GenomeAgricultural Genomics Institute at ShenzhenChinese Academy of Agricultural SciencesShenzhen518124China
- Kunpeng Institute of Modern Agriculture at FoshanChinese Academy of Agricultural SciencesFoshan528226China
| |
Collapse
|
32
|
He G, Yao H, Duan S, Luo L, Sun Q, Tang R, Chen J, Wang Z, Sun Y, Li X, Hu L, Yun L, Yang J, Yan J, Nie S, Zhu Y, Wang CC, Liu B, Hu L, Liu C, Wang M. Pilot work of the 10K Chinese People Genomic Diversity Project along the Silk Road suggests a complex east-west admixture landscape and biological adaptations. SCIENCE CHINA. LIFE SCIENCES 2025; 68:914-933. [PMID: 39862346 DOI: 10.1007/s11427-024-2748-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/27/2024] [Indexed: 01/27/2025]
Abstract
Genomic sources from China are underrepresented in the population-specific reference database. We performed whole-genome sequencing or genome-wide genotyping on 1,207 individuals from four linguistically diverse groups (1,081 Sinitic, 56 Mongolic, 40 Turkic, and 30 Tibeto-Burman people) living in North China included in the 10K Chinese People Genomic Diversity Project (10K_CPGDP) to characterize the genetic architecture and adaptative history of ethnic groups in the Silk Road Region of China. We observed a population split between Northwest Chinese minorities (NWCMs) and Han Chinese since the Upper Paleolithic and later Neolithic genetic differentiation within NWCMs. The observed population substructures among ethnically/linguistically diverse NWCMs suggested that differentiated admixture events contributed to the differences in their genomic and phenotypic diversity. We estimated that the Dongxiang, Tibetan, and Yugur people inherited more than 10% of the Western Eurasian ancestry, which is much greater than that of the Salar and Tu people (<7%), while Han neighbors showed less West Eurasian ancestry (∼1%-3%). Male-biased admixture introduced Western Eurasian ancestry in the Dongxiang, Tibetan, and Yugur populations. We found that the eastern-western admixture in NWCMs occurred ∼800-1,100 years ago, coinciding with intensive economic and cultural exchanges during the Tang and Song dynasties. Additionally, we identified the signatures of natural selection associated with cardiovascular system diseases or lipid metabolism and developmental/neurogenetic disorders. Moreover, the EPAS1 gene showed relatively high population branch statistic values in NWCMs. The well-fitted demographical models presented the vast landscape of complex admixture processes of the Silk Road people, and the newly reported functionally important variations suggested the importance of including ethnolinguistically diverse populations in Chinese genetic studies for uncovering the genetic basis of complex traits/diseases.
Collapse
Affiliation(s)
- Guanglin He
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China.
- Center for Archaeological Science, Sichuan University, Chengdu, 610000, China.
| | - Hongbing Yao
- Belt and Road Research Center for Forensic Molecular Anthropology, Gansu University of Political Science and Law, Lanzhou, 730000, China
| | - Shuhan Duan
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- School of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637007, China
- Center for Genetics and Prenatal Diagnosis, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637007, China
- Institution of Genome Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637007, China
| | - Lintao Luo
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, 400331, China
| | - Qiuxia Sun
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, 400331, China
| | - Renkuan Tang
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, 400331, China
| | - Jing Chen
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030001, China
| | - Zhiyong Wang
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Yuntao Sun
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- West China School of Basic Science & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiangping Li
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Liping Hu
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Libing Yun
- West China School of Basic Science & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Junbao Yang
- School of Basic Medicine and Forensic Medicine, North Sichuan Medical College, Nanchong, 637007, China
- Center for Genetics and Prenatal Diagnosis, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637007, China
- Institution of Genome Medicine, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637007, China
| | - Jiangwei Yan
- School of Forensic Medicine, Shanxi Medical University, Jinzhong, 030001, China
| | - Shengjie Nie
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Yanfeng Zhu
- Department of Public Health, Chengdu Medical College, Chengdu, 610500, China
| | - Chuan-Chao Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, 361005, China
| | - Bing Liu
- Institute of Forensic Science, Ministry of Public Security, Beijing, 100038, China
| | - Lan Hu
- Institute of Forensic Science, Ministry of Public Security, Beijing, 100038, China
| | - Chao Liu
- Anti-Drug Technology Center of Guangdong Province, Guangzhou, 510220, China.
| | - Mengge Wang
- Institute of Rare Diseases, West China Hospital of Sichuan University, Sichuan University, Chengdu, 610000, China.
- Center for Archaeological Science, Sichuan University, Chengdu, 610000, China.
- Anti-Drug Technology Center of Guangdong Province, Guangzhou, 510220, China.
- Department of Forensic Medicine, College of Basic Medicine, Chongqing Medical University, Chongqing, 400331, China.
| |
Collapse
|
33
|
Cousins T, Scally A, Durbin R. A structured coalescent model reveals deep ancestral structure shared by all modern humans. Nat Genet 2025; 57:856-864. [PMID: 40102687 PMCID: PMC11985351 DOI: 10.1038/s41588-025-02117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025]
Abstract
Understanding the history of admixture events and population size changes leading to modern humans is central to human evolutionary genetics. Here we introduce a coalescence-based hidden Markov model, cobraa, that explicitly represents an ancestral population split and rejoin, and demonstrate its application on simulated and real data across multiple species. Using cobraa, we present evidence for an extended period of structure in the history of all modern humans, in which two ancestral populations that diverged ~1.5 million years ago came together in an admixture event ~300 thousand years ago, in a ratio of ~80:20%. Immediately after their divergence, we detect a strong bottleneck in the major ancestral population. We inferred regions of the present-day genome derived from each ancestral population, finding that material from the minority correlates strongly with distance to coding sequence, suggesting it was deleterious against the majority background. Moreover, we found a strong correlation between regions of majority ancestry and human-Neanderthal or human-Denisovan divergence, suggesting the majority population was also ancestral to those archaic humans.
Collapse
Affiliation(s)
- Trevor Cousins
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Aylwyn Scally
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Richard Durbin
- Department of Genetics, University of Cambridge, Cambridge, UK.
| |
Collapse
|
34
|
Patin E, Quintana-Murci L. Tracing the Evolution of Human Immunity Through Ancient DNA. Annu Rev Immunol 2025; 43:57-82. [PMID: 39705165 DOI: 10.1146/annurev-immunol-082323-024638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
Infections have imposed strong selection pressures throughout human evolution, making the study of natural selection's effects on immunity genes highly complementary to disease-focused research. This review discusses how ancient DNA studies, which have revolutionized evolutionary genetics, increase our understanding of the evolution of human immunity. These studies have shown that interbreeding between modern humans and Neanderthals or Denisovans has influenced present-day immune responses, particularly to viruses. Additionally, ancient genomics enables the tracking of how human immunity has evolved across cultural transitions, highlighting strong selection since the Bronze Age in Europe (<4,500 years) and potential genetic adaptations to epidemics raging during the Middle Ages and the European colonization of the Americas. Furthermore, ancient genomic studies suggest that the genetic risk for noninfectious immune disorders has gradually increased over millennia because alleles associated with increased risk for autoimmunity and inflammation once conferred resistance to infections. The challenge now is to extend these findings to diverse, non-European populations and to provide a more global understanding of the evolution of human immunity.
Collapse
Affiliation(s)
- Etienne Patin
- Institut Pasteur, Université Paris Cité, CNRS UMR 2000, Human Evolutionary Genetics Unit, Paris, France;
| | - Lluis Quintana-Murci
- Human Genomics and Evolution, Collège de France, Paris, France
- Institut Pasteur, Université Paris Cité, CNRS UMR 2000, Human Evolutionary Genetics Unit, Paris, France;
| |
Collapse
|
35
|
Ruiz-Ramírez J, Bittner F, Parsons TJ, Tillmar A, Vangeel L, Grandell I, Eduardoff M, Peck MA, Ambroa-Conde A, Mosquera-Miguel A, Freire-Aradas A, Lareu MV, Phillips C, de la Puente M. Inter-platform evaluation of the MPSplex large-scale tri-allelic SNP panel for forensic identification. Forensic Sci Int Genet 2025; 77:103233. [PMID: 40037007 DOI: 10.1016/j.fsigen.2025.103233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 03/06/2025]
Abstract
MPSplex is a large-scale forensic massively parallel sequencing (MPS) panel with 1,270 tri-allelic SNPs, 44 microhaplotypes (MH) and 55 ancestry-informative bi-allelic SNPs (aiSNPs) designed for missing persons identification. We have evaluated MPSplex with the most widely used MPS platforms in the forensic field: the Illumina MiSeq, the Thermo Fisher Scientific Ion S5 and the Qiagen GeneReader. The tri-allelic SNPs of MPSplex were previously identified from the most polymorphic loci with three common alleles in 1000 Genomes Phase III data and combined with the 44 MH and 55 aiSNPs, then implemented into a QIAseq Targeted DNA Custom Panel (Qiagen), a marker panel which uses Unique Molecular Indices or UMIs. The UMI random-sequence DNA molecules are incorporated onto DNA fragments before the Target Enrichment PCR, allowing the identification of reads that originated from the same template and consequently they can be used to correct the errors that may arise within the PCR or the sequencing process. In this study, we present the results of an inter-platform evaluation of the MPSplex panel, characterizing its performance in different forensic scenarios, which assessed aspects that include sensitivity, genotyping accuracy and mixture analysis. MPSplex aims to provide a tool designed for kinship analysis that can be applied beyond the resolution of first- or second-degree relationships, avoiding the need for much bigger forensic panels designed for genealogy purposes, which usually require significantly more sequencing resources. This study provides evaluation of MPSplex using the MPS systems in routine use for forensic genotyping of large-scale panels of SNPs.
Collapse
Affiliation(s)
- J Ruiz-Ramírez
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
| | - F Bittner
- International Commission on Missing Persons, Koninginnegracht 12, The Hague, Netherlands
| | - T J Parsons
- International Commission on Missing Persons, Koninginnegracht 12, The Hague, Netherlands
| | - A Tillmar
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden; Department of Biomedical and Clinical Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - L Vangeel
- International Commission on Missing Persons, Koninginnegracht 12, The Hague, Netherlands
| | - I Grandell
- Department of Forensic Genetics and Forensic Toxicology, National Board of Forensic Medicine, Linköping, Sweden
| | - M Eduardoff
- International Commission on Missing Persons, Koninginnegracht 12, The Hague, Netherlands
| | - M A Peck
- International Commission on Missing Persons, Koninginnegracht 12, The Hague, Netherlands
| | - A Ambroa-Conde
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - A Mosquera-Miguel
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - A Freire-Aradas
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - M V Lareu
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - C Phillips
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Santiago de Compostela, Spain; King's Forensics, Faculty of Life Sciences and Medicine, King's College, London, UK
| | - M de la Puente
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Santiago de Compostela, Spain.
| |
Collapse
|
36
|
Zhu H, Wang Y, Li L, Wang L, Zhang H, Jin X. Cell-free DNA from clinical testing as a resource of population genetic analysis. Trends Genet 2025; 41:330-344. [PMID: 39578178 DOI: 10.1016/j.tig.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
As a noninvasive biomarker, cell-free DNA (cfDNA) has achieved remarkable success in clinical applications. Notably, cfDNA is essentially DNA, and conducting whole-genome sequencing (WGS) can yield a wealth of genetic information. These invaluable data should not be confined to one-time use; instead, they should be leveraged for more comprehensive population genetic analysis, including genetic variation spectrum, population structure and genetic selection, and genome-wide association studies (GWASs), among others. Such research findings can, in turn, facilitate clinical practice, enabling more advanced and accurate disease predictions. This review explores the advantages, challenges, and current research areas of cfDNA in population genetics. We hope that this review can serve as a new chapter in the repurposing of cfDNA sequence data generated from clinical testing in population genetics.
Collapse
Affiliation(s)
- Huanhuan Zhu
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Yu Wang
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Linxuan Li
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China; College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Wang
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Haiqiang Zhang
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China
| | - Xin Jin
- BGI Research, Shenzhen 518083, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China; School of Medicine, South China University of Technology, Guangzhou 510641, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
37
|
Wang Y, Sams EI, Slaugh R, Crocker S, Hurtado EC, Tracy S, Hou YCC, Markovic C, Valle K, Tate V, Belhassan K, Appelbaum E, Akinwe T, Tzovenos RS, Cao Y, Neilson A, Liu Y, Jensen N, Ghasemi R, Lindsay T, Manuel J, Couteranis S, Kremitzki M, Ustanik J, Antonacci T, Ng JK, Emory A, Metz L, DeLuca T, Lyons KN, Sinnwell T, Thomeczek B, Wang K, Sisneros N, Muraleedharan M, Kethireddy A, Corbo M, Gowda H, King K, Gurnett CA, Dutcher SK, Gooch C, Li YE, Mitchell MW, Peterson KA, Horani A, Rosenfeld JA, Bi W, Stankiewicz P, Chao HT, Posey J, Grochowski CM, Dardas Z, Puffenberger E, Pearson CE, Kooy F, Annear D, Innes AM, Heinz M, Head R, Fulton R, Toutain S, 9P-ARCH, Antonacci-Fulton L, Cui X, Mitra RD, Cole FS, Neidich J, Dickson PI, Milbrandt J, Turner TN. Whole-Genome Sequencing Reveals Individual and Cohort Level Insights into Chromosome 9p Syndromes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.28.25324850. [PMID: 40196253 PMCID: PMC11974940 DOI: 10.1101/2025.03.28.25324850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Previous genomic efforts on chromosome 9p deletion and duplication syndromes have utilized low resolution strategies (i.e., karyotypes, chromosome microarrays). We present the first large-scale whole-genome sequencing (WGS) study of 100 individuals from families with 9p-related syndromes including 85 unrelated probands through the 9P-ARCH (Advanced Research in Chromosomal Health: Genomic, Phenotypic, and Functional Aspects of 9p-Related syndromes) research network. We analyzed the genomic architecture of these syndromes, highlighting fundamental features and their commonalities and differences across individuals. This work includes a machine-learning model that predicts 9p deletion syndrome from gene copy number estimates using WGS data. Two Late Replicating Regions (LRR1 [a previously un-named human fragile site], LRR2) were identified that contain most structural variant breakpoints in 9p deletion syndrome pointing to replication-based issues in structural variant formation. Furthermore, we show the utility of using WGS information to obtain a comprehensive understanding of 9p-related variation in an individual with complex structural variation where chromothripsis is the likely mechanism. Genes on 9p were prioritized based on statistical assessment of human genomic variation. Furthermore, through application of spatial transcriptomics to embryonic mouse tissue we examined 9p-gene expression in craniofacial and brain development. Through these strategies, we identified 24 important genes for the majority (83%) of individuals with 9p deletion syndrome including AK3, BRD10, CD274, CDC37L1, DMRT1, DMRT2, DMRT3, DOCK8, GLIS3, JAK2, KANK1, KDM4C, PLPP6, PTPRD, PUM3, RANBP6, RCL1, RFX3, RIC1, SLC1A1, SMARCA2, UHRF2, VLDLR, and ZNG1A. Two genes (AK3, ZNG1A) are involved in mitochondrial function and testing of the mitochondrial genome revealed excess copy number in individuals with 9p deletion syndrome. This study presents the most comprehensive genomic analysis of 9p-related syndromes to date, with plans for further expansion through our 9P-ARCH research network.
Collapse
Affiliation(s)
- Yingxi Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eleanor I. Sams
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rachel Slaugh
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sandra Crocker
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Emily Cordova Hurtado
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sophia Tracy
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ying-Chen Claire Hou
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher Markovic
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kostandin Valle
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Victoria Tate
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Khadija Belhassan
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Elizabeth Appelbaum
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Titilope Akinwe
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
- Washington University Pain Center, Dept. of Anesthesiology, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Rodrigo Starosta Tzovenos
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yang Cao
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amber Neilson
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yu Liu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nathaniel Jensen
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Reza Ghasemi
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tina Lindsay
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Juana Manuel
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sophia Couteranis
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Milinn Kremitzki
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jack Ustanik
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Thomas Antonacci
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey K. Ng
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrew Emory
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laura Metz
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tracie DeLuca
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Katherine N. Lyons
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Toni Sinnwell
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brianne Thomeczek
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | - Marco Corbo
- Medgenome Laboratory, Foster City, CA 94404, USA
| | - Harsha Gowda
- Medgenome Laboratory, Foster City, CA 94404, USA
| | - Katherine King
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christina A. Gurnett
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan K. Dutcher
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Catherine Gooch
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yang E. Li
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Neurosugery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | - Amjad Horani
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jill A. Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Baylor Genetics Laboratory, Baylor College of Medicine, Houston, TX 77030, USA
| | - Weimin Bi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Baylor Genetics Laboratory, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pawel Stankiewicz
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hsiao-Tuan Chao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jennifer Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christopher M. Grochowski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zain Dardas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Frank Kooy
- Department of Medical Genetics, University of Antwerp, Edegem, Belgium
| | - Dale Annear
- Department of Medical Genetics, University of Antwerp, Edegem, Belgium
| | - A. Micheil Innes
- Departments of Medical Genetics and Pediatrics, Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael Heinz
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Richard Head
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert Fulton
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - 9P-ARCH
- 9P-ARCH Research Network - Advanced Research in Chromosomal Health: Genomic, Phenotypic, and Functional Aspects of 9p-Related syndromes
| | | | - Xiaoxia Cui
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robi D. Mitra
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - F. Sessions Cole
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Julie Neidich
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Patricia I. Dickson
- Edward Mallinckrodt Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63110, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, St. Louis, MO, USA
| | - Tychele N. Turner
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
38
|
Nauseef JT, Chu TR, Hooper WF, Alonso A, Oku A, Geiger H, Goldstein ZR, Shah M, Sigouros M, Manohar J, Steinsnyder Z, Winterkorn L, Robinson BD, Sboner A, Beltran H, Elemento O, Hajirasouliha I, Imielinski M, Nanus DM, Tagawa ST, Robine N, Mosquera JM. A complex phylogeny of lineage plasticity in metastatic castration resistant prostate cancer. NPJ Precis Oncol 2025; 9:91. [PMID: 40155466 PMCID: PMC11953479 DOI: 10.1038/s41698-025-00854-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 02/25/2025] [Indexed: 04/01/2025] Open
Abstract
Aggressive variant and androgen receptor (AR)-independent castration resistant prostate cancers (CRPC) represent the most significant diagnostic and therapeutic challenges in prostate cancer. This study examined a case of simultaneous progression of both adenocarcinoma and squamous tumors from the same common origin. Using whole-genome and transcriptome sequencing from 17 samples collected over >6 years, we established the clonal relationship of all samples, defined shared complex structural variants, and demonstrated both divergent and convergent evolution at AR. Squamous CRPC-associated circulating tumor DNA was identified at clinical progression prior to biopsy detection of any squamous differentiation. Dynamic changes in the detection rate of histology-specific clones in circulation reflected histology-specific sensitivity to treatment. This dataset serves as an illustration of non-neuroendocrine transdifferentiation and highlights the importance of serial sampling at progression in CRPC for the detection of emergent non-adenocarcinoma histologies with implications for the treatment of lineage plasticity and transdifferentiation in metastatic CRPC.
Collapse
Affiliation(s)
- Jones T Nauseef
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, NY, USA.
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
- New York Genome Center, New York, NY, USA.
| | | | | | - Alicia Alonso
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ali Oku
- New York Genome Center, New York, NY, USA
| | | | | | | | - Michael Sigouros
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jyothi Manohar
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | | | - Brian D Robinson
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Andrea Sboner
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Himisha Beltran
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, New York, NY, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Iman Hajirasouliha
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Marcin Imielinski
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- New York Genome Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - David M Nanus
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Scott T Tagawa
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | | | - Juan Miguel Mosquera
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA.
- New York Genome Center, New York, NY, USA.
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
39
|
Gravel S. Mapping a complex evolutionary history. Science 2025; 387:1352-1353. [PMID: 40146848 DOI: 10.1126/science.adw5484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Tracking the geographic origins of genetic ancestors reveals past human migrations.
Collapse
Affiliation(s)
- Simon Gravel
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| |
Collapse
|
40
|
Luo T, Guo W, Ji W, Du W, Lv Y, Feng Z. Monocyte CCL2 signaling possibly contributes to increased asthma susceptibility in type 2 diabetes. Sci Rep 2025; 15:10768. [PMID: 40155667 PMCID: PMC11953320 DOI: 10.1038/s41598-025-95039-1] [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] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
In recent years, the respiratory system has been increasingly recognized as a key target organ in diabetes. Although observational studies have established significant clinical associations between type 2 diabetes (T2D), antidiabetic medication use, and asthma, the causal relationships and underlying molecular mechanisms remain unclear. This study employed a bidirectional two-sample Mendelian randomization (MR) approach combined with bioinformatics analysis to explore the causal relationships between T2D and asthma subtypes and complications, with a focus on immune-regulatory mechanisms. The MR analysis utilized inverse-variance weighted (IVW) and meta-analysis methods to evaluate overall effects, with sensitivity analyses confirming the robustness of the findings. Bioinformatics analysis focused on differential gene expression and pathway enrichment to identify potential molecular networks. The MR analysis showed that T2D has a significant positive causal effect on asthma (P < 0.05), with severe autoimmune T2D showing strong associations with specific asthma subtypes (eosinophilic and mixed asthma) and complications (e.g., acute respiratory infections and pneumonia) (P < 0.05). Bioinformatics analysis identified the monocyte-CCL2 signaling axis as a key mechanism linking T2D and asthma, where hyperglycemia-induced monocyte activation may promote asthma development. These findings reveal shared inflammatory pathways and deepen our understanding of the molecular mechanisms linking these two chronic diseases.
Collapse
Affiliation(s)
- Tian Luo
- Department of Respiratory and Critical Care Medicine, The People's Hospital of Sishui, Jining, 273200, Shandong, China
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Weihong Guo
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Wentao Ji
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - WeiWei Du
- Zhongshan City People's Hospital, Xinxiang Medical University, Xinxiang, 453003, Henan, China
| | - Yanhua Lv
- Department of Respiratory and Critical Care Medicine, Shunde Hospital of Southern Medical University, Shunde, 528300, Guangdong, China.
| | - Zhijun Feng
- Postdoctoral Innovation Practice Base, Jiangmen Central Hospital, Southern Medical University, Jiangmen, 529030, Guangdong, China.
| |
Collapse
|
41
|
Cisternino F, Song Y, Peters TS, Westerman R, de Borst GJ, Diez Benavente E, van den Dungen NA, Homoed-van der Kraak P, de Kleijn DP, Mekke J, Mokry M, Pasterkamp G, den Ruijter HM, Velema E, Miller CL, Glastonbury CA, van der Laan S. Intraplaque haemorrhage quantification and molecular characterisation using attention based multiple instance learning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.04.25323316. [PMID: 40093230 PMCID: PMC11908327 DOI: 10.1101/2025.03.04.25323316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Intraplaque haemorrhage (IPH) represents a critical feature of plaque vulnerability as it is robustly associated with adverse cardiovascular events, including stroke and myocardial infarction. How IPH drives plaque instability is unknown. However, its identification and quantification in atherosclerotic plaques is currently performed manually, with high interobserver variability, limiting its accurate assessment in large cohorts. Leveraging the Athero-Express biobank, an ongoing study comprising a comprehensive dataset of histological, transcriptional, and clinical information from 2,595 carotid endarterectomy patients, we developed an attention-based additive multiple instance learning (MIL) framework to automate the detection and quantification of IPH across whole-slide images of nine distinct histological stains. We demonstrate that routinely available Haematoxylin and Eosin (H&E) staining outperformed all other plaque relevant Immunohistochemistry (IHC) stains tested (AUROC = 0.86), underscoring its utility in quantifying IPH. When combining stains through ensemble models, we see that H&E + CD68 (a macrophage marker) as well as H&E + Verhoeff-Van Gieson elastic fibers staining (EVG) leads to a substantial improvement (AUROC = 0.92). Using our model, we could derive IPH area from the MIL-derived patch-level attention scores, enabling not only classification but precise localisation and quantification of IPH area in each plaque, facilitating downstream analyses of its association and cellular composition with clinical outcomes. By doing so, we demonstrate that IPH presence and area are the most significant predictors of both preoperative symptom presentation and major adverse cardiovascular events (MACE), outperforming manual scoring methods. Automating IPH detection also allowed us to characterise IPH on a molecular level at scale. Pairing IPH measurements with single-cell transcriptomic analyses revealed key molecular pathways involved in IPH, including TNF-α signalling, extracellular matrix remodelling and the presence of foam cells. This study represents the largest effort in the cardiovascular field to integrate digital pathology, machine learning, and molecular data to predict and characterize IPH which leads to better understanding how it drives symptoms and MACE. Our model provides a scalable, interpretable, and reproducible method for plaque phenotyping, enabling the derivation of plaque phenotypes for predictive modelling of MACE outcomes.
Collapse
Affiliation(s)
| | - Yipei Song
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Computer Engineering, University of Virginia, Charlottesville, VA, USA
| | - Tim S. Peters
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Roderick Westerman
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gert J. de Borst
- Vascular surgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ernest Diez Benavente
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Noortje A.M. van den Dungen
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Dominique P.V. de Kleijn
- Vascular surgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Joost Mekke
- Vascular surgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Michal Mokry
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Hester M. den Ruijter
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Experimental Cardiology, Department Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Evelyn Velema
- Experimental Cardiology, Department Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Clint L. Miller
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Craig A. Glastonbury
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157, Milan, Italy
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - S.W. van der Laan
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
42
|
Rao J, Luo H, An D, Liang X, Peng L, Chen F. Performance evaluation of structural variation detection using DNBSEQ whole-genome sequencing. BMC Genomics 2025; 26:299. [PMID: 40133825 PMCID: PMC11938577 DOI: 10.1186/s12864-025-11494-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND DNBSEQ platforms have been widely used for variation detection, including single-nucleotide variants (SNVs) and short insertions and deletions (INDELs), which is comparable to Illumina. However, the performance and even characteristics of structural variations (SVs) detection using DNBSEQ platforms are still unclear. RESULTS In this study, we assessed the detection of SVs using 40 tools on eight DNBSEQ whole-genome sequencing (WGS) datasets and two Illumina WGS datasets of NA12878. Our findings confirmed that the performance of SVs detection using the same tool on DNBSEQ and Illumina datasets was highly consistent, with correlations greater than 0.80 on metrics of number, size, precision and sensitivity, respectively. Furthermore, we constructed a "DNBSEQ" SV set (4,785 SVs) from the DNBSEQ datasets and an "Illumina" SV set (6,797 SVs) from the Illumina datasets. We found that these two SV sets were highly consistent of SV sites and genomic characteristics, including repetitive regions, GC distribution, difficult-to-sequence regions, and gene features, indicating the robustness of our comparative analysis and highlights the value of both platforms in understanding the genomic context of SVs. CONCLUSIONS Our study systematically analyzed and characterized germline SVs detected on WGS datasets sequenced from DNBSEQ platforms, providing a benchmark resource for further studies of SVs using DNBSEQ platforms.
Collapse
Affiliation(s)
- Junhua Rao
- MGI Tech, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | | | - Dan An
- MGI Tech, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xinming Liang
- MGI Tech, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | | | - Fang Chen
- MGI Tech, Shenzhen, 518083, China.
- BGI, Shenzhen, 518083, China.
| |
Collapse
|
43
|
Yang Y, Chen J, Zhao X, Gong F, Liu R, Miao J, Lin M, Ge F, Chen W. Genetic analysis reveals the shared genetic architecture between breast cancer and atrial fibrillation. Front Genet 2025; 16:1450259. [PMID: 40201568 PMCID: PMC11975938 DOI: 10.3389/fgene.2025.1450259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 02/28/2025] [Indexed: 04/10/2025] Open
Abstract
Background Epidemiological studies have observed an association between atrial fibrillation (AF) and breast cancer (BC). However, the underlying mechanisms linking these two conditions remain unclear. This study aims to systematically explore the genetic association between AF and BC. Methods We utilized the largest available genome-wide association study (GWAS) datasets for European individuals, including summary data for AF (N = 1,030,836) and BC (N = 247,173). Multiple approaches were employed to systematically investigate the genetic relationship between AF and BC from the perspectives of pleiotropy and causality. Results Global genetic analysis using LDSC and HDL revealed a genetic correlation between AF and BC (rg = 0.0435, P = 0.039). Mixer predicted genetic overlap between non-MHC regions of the two conditions (n = 125, rg = 0.05). Local genetic analyses using LAVA and GWAS-PW identified 22 regions with potential genetic sharing. Cross-trait meta-analysis by CPASSOC identified one novel pleiotropic SNP and 14 pleiotropic SNPs, which were subsequently annotated. Eight of these SNPs passed Bayesian colocalization tests, including one novel pleiotropic SNP. Further fine-mapping analysis identified a set of causal SNPs for each significant SNP. TWAS analyses using JTI and FOCUS models jointly identified 10 pleiotropic genes. Phenome-wide association study (PheWAS) of novel pleiotropic SNPs identified two eQTLs (PELO, ITGA1). Gene-based PheWAS results showed strong associations with BMI, height, and educational attainment. PCGA methods combining GTEx V8 tissue data and single-cell RNA data identified 16 co-enriched tissue types (including cardiovascular, reproductive, and digestive systems) and 5 cell types (including macrophages and smooth muscle cells). Finally, univariable and multivariable bidirectional Mendelian randomization analyses excluded a causal relationship between AF and BC. Conclusion This study systematically investigated the shared genetic overlap between AF and BC. Several pleiotropic SNPs and genes were identified, and co-enriched tissue and cell types were revealed. The findings highlight common mechanisms from a genetic perspective rather than a causal relationship. This study provides new insights into the AF-BC association and suggests potential experimental targets and directions for future research. Additionally, the results underscore the importance of monitoring the potential risk of one disease in patients diagnosed with the other.
Collapse
Affiliation(s)
- Yang Yang
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, China
| | - Jiayi Chen
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, China
| | - XiaoHua Zhao
- Department of Cardiology, Yan’an Hospital Affiliated To Kunming Medical University, Kunming, China
| | - Fuhong Gong
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, China
| | - Ruimin Liu
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, China
| | - Jingge Miao
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, China
| | - Mengping Lin
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, China
| | - Fei Ge
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Wenlin Chen
- Yunnan Key Laboratory of Breast Cancer Precision Medicine, Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Yunnan Cancer Hospital, Kunming, China
| |
Collapse
|
44
|
Chukwu W, Lee S, Crane A, Zhang S, Webster S, Dakhama O, Mittra I, Rauert C, Imielinski M, Beroukhim R, Dubois F, Dalin S. A sequence context-based approach for classifying tumor structural variants without paired normal samples. CELL REPORTS METHODS 2025; 5:100991. [PMID: 40081367 PMCID: PMC12049684 DOI: 10.1016/j.crmeth.2025.100991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/13/2024] [Accepted: 02/12/2025] [Indexed: 03/16/2025]
Abstract
Although several recent studies have characterized structural variants (SVs) in germline and cancer genomes independently, the genomic contexts of these SVs have not been comprehensively compared. We examined similarities and differences between 2 million germline and 115 thousand tumor SVs from a cohort of 963 patients from The Cancer Genome Atlas. We found significant differences in features related to their genomic sequences and localization that suggest differences between SV-generating processes and selective pressures. For example, our results show that features linked to transposon-mediated processes are associated with germline SVs, while somatic SVs more frequently show features characteristic of chromoanagenesis. These genomic differences enabled us to develop a classifier-the Germline and Tumor Structural Variant or "the great GaTSV" -that accurately distinguishes between germline and cancer SVs in tumor samples that lack a matched normal sample.
Collapse
Affiliation(s)
- Wolu Chukwu
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Siyun Lee
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alexander Crane
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shu Zhang
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sophie Webster
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Oumayma Dakhama
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ipsa Mittra
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carlos Rauert
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany
| | - Marcin Imielinski
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA; New York Genome Center, New York, NY, USA; Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA; Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA; Department of Pathology and Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Rameen Beroukhim
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Frank Dubois
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Institute of Pathology, Berlin, Germany.
| | - Simona Dalin
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
45
|
Wei G, Chen R, Liu S, Cai S, Feng Z. Telomere Length as Both Cause and Consequence in Type 1 Diabetes: Evidence from Bidirectional Mendelian Randomization. Biomedicines 2025; 13:774. [PMID: 40299325 PMCID: PMC12024553 DOI: 10.3390/biomedicines13040774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
Background/Objectives: Diabetes is the most prevalent metabolic disease globally, characterized by dysregulated glucose control and accompanied by multiple refractory complications. As a critical marker of cellular homeostasis, telomere length (TL) may be associated with the progression of diabetes. However, the causal relationship between diabetes and TL remains unclear, particularly whether cellular homeostasis imbalance acts as a consequence of diabetic complications or a precipitating factor in disease development. Methods: We performed a bidirectional Mendelian randomization (MR) analysis using genome-wide association study (GWAS) data. Following the three core assumptions of MR analysis, we conducted quality control on all instrumental variables to ensure methodological rigor. The inverse variance weighted (IVW) method served as the primary analytical method, supplemented by additional MR methods to evaluate the significance of the results. Furthermore, we performed sensitivity analyses to ensure the reliability and robustness of the findings. Results: Forward analysis revealed that shortened TL significantly increases the risk of broadly defined Type 1 diabetes (T1D) and unspecified types of diabetes (p < 0.05). Additionally, we identified a positive causal relationship between TL and several diabetes-related complications, including co-morbidities, diabetic nephropathy, and diabetic ketoacidosis (p < 0.05). Interestingly, the reverse analysis demonstrated a positive causal effect of T1D and its complications on TL (p < 0.05); however, this effect disappeared after adjusting for insulin use (p > 0.05). Conclusions: Bidirectional MR analyses revealed a complex relationship between TL and T1D, where shortened telomeres increase T1D risk while T1D itself may trigger compensatory mechanisms affecting telomere maintenance, with insulin playing a crucial regulatory role in this relationship. These findings suggest telomere biology may be fundamentally involved in T1D pathogenesis and could inform future therapeutic approaches.
Collapse
Affiliation(s)
- Guanping Wei
- Department of Emergency, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Ruiping Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Shupeng Liu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China;
| | - Shenhua Cai
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhijun Feng
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China;
| |
Collapse
|
46
|
Xiao J, Feng C, Zhu T, Zhang X, Chen X, Li Z, You J, Wang Q, Zhuansun D, Meng X, Wang J, Xiang L, Yu X, Zhou B, Tang W, Tou J, Wang Y, Yang H, Yu L, Liu Y, Jiang X, Ren H, Yu M, Chen Q, Yin Q, Liu X, Xu Z, Wu D, Yu D, Wu X, Yang J, Xiong B, Chen F, Hao X, Feng J. Rare and common genetic variants underlying the risk of Hirschsprung's disease. Hum Mol Genet 2025; 34:586-598. [PMID: 39817569 DOI: 10.1093/hmg/ddae205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/18/2025] Open
Abstract
Hirschsprung's disease (HSCR) is a congenital enteric neuropathic disorder characterized by high heritability (>80%) and polygenic inheritance (>20 genes). The previous genome-wide association studies (GWAS) identified several common variants associated with HSCR and demonstrated increased predictive performance for HSCR risk in Europeans using a genetic risk score, there remains a notable gap in knowledge regarding Chinese populations. We conducted whole exome sequencing in a HSCR case cohort in Chinese. By using the common controls (505 controls from 1KG EAS and 10 588 controls from ChinaMAP), we conducted GWAS for the common variants in the exome and gene-based association for rare variants. We further validated the associated variants and genes in replicated samples and in vitro and vivo experiments. We identified one novel gene PLK5 by GWAS and suggested 45 novel putative genes based the gene-based test. By using genetic variant at RET and PLK5, we constructed a genetic risk score that could identify the individuals with very high genetic risk for HSCR. Compared with patients with zero or one risk allele from the three variants, the risk for HSCR was 36.61 times higher with six alleles. In addition, we delineated a HSCR risk gene landscape that encompasses 57 genes, which explains 88.5% and 54.5% of HSCR in Chinese and European, respectively. In summary, this study improved the understanding of genetic architecture of HSCR and provided a risk prediction approach for HSCR in the Chinese.
Collapse
Affiliation(s)
- Jun Xiao
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Chenzhao Feng
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Tianqi Zhu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Xuan Zhang
- Department of Pediatric Surgery, Pingshan District Maternal & Child Healthcare Hospital of Shenzhen, No. 6 Longtian South Road, Longtian Subdistrict, Pingshan District, Shenzhen, Guangdong 518122, China
| | - Xuyong Chen
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Zejian Li
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Jingyi You
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Qiong Wang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Didi Zhuansun
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Xinyao Meng
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Jing Wang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Lei Xiang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Xiaosi Yu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Bingyan Zhou
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Weibing Tang
- Department of Pediatric Surgery, Children's Hospital of Nanjing Medical University, No. 72 Guangzhou Road, Gulou District, Nanjing, Jiangsu 210008, China
| | - Jinfa Tou
- Department of General Surgery, Children's Hospital, Zhejiang University School of Medicine, No. 3333 Binsheng Road, Binjiang District, Hangzhou, Zhejiang 310003, China
| | - Yi Wang
- Department of General and Neonatal Surgery, Children's Hospital of Chongqing Medical University, No. 136, Zhongshan 2nd Road, Yuzhong District, Chongqing 400014, China
| | - Heying Yang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, No. 1 Renmin Road, Erqi District, Henan 450052, China
| | - Lei Yu
- Department of Neonatal Surgery, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 100 Hong Kong Road, Jiang'an District, Wuhan, Hubei 430030, China
| | - Yuanmei Liu
- Department of Pediatric Surgery, The Affiliated Hospital of Zunyi Medical University, No. 149 Dalian Road, Huichuan District, Zunyi, Guizhou 563000, China
| | - Xuewu Jiang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Shantou University Medical College, No. 69, Dongxia North Road, Jinping District, Shantou, Guangdong 515041, China
| | - Hongxia Ren
- Department of Neonatal Surgery, Children's Hospital of Shanxi, No. 13 Xinminbei Street, Xinhualing district, Taiyuan, Shanxi 030013, China
| | - Mei Yu
- Department of Pediatric Surgery, Guiyang Maternal and Child Health Hospital, No. 63 Ruijin South Road, Nanming district, Guiyang, Guizhou 550002, China
| | - Qi Chen
- Department of Pediatric Surgery, The Third Affiliated Hospital of Zhengzhou University, No. 7 Kangfuqian Street, Erqi District, Zhengzhou 450052, Henan, China
| | - Qiang Yin
- Department of General Surgery, Hunan Children's Hospital, No. 86 Ziyuan Road, Yuhua District, Changsha, Hunan 515041, China
| | - Xiang Liu
- Department of Pediatric Surgery, Anhui Provincial Children's Hospital, No. 39 Wangjiang East Road, Wuhu Road Subdistrict, Hefei, Anhui 230051, China
| | - Zhilin Xu
- Department of Pediatric Surgery, The First Affiliated Hospital of Harbin Medical University, No. 199 Dazhi Street, Nangang district, Harbin, Heilongjiang 150001, China
| | - Dianming Wu
- Department of Pediatric Surgery, Fujian Maternity and Child Health Hospital, Fujian Medical University, No. 18 Daoshan Road, Gulou District, Fuzhou, Fujian 350001, China
| | - Donghai Yu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Xiaojuan Wu
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Jixin Yang
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| | - Bo Xiong
- Department of Forensic Medicine, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Qiaokou District, Wuhan, Hubei 430030, China
| | - Feng Chen
- Department of Pediatric Surgery, Union Hospital, Fujian Medical University, No. 29, Xinquan Road, Gulou District, Fuzhou, Fujian 350001, China
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College Huazhong University of Science and Technology, No. 13 Hangkong Road, Qiaokou District, Wuhan, Hubei 430030, China
| | - Jiexiong Feng
- Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
- Hubei Clinical Center of Hirschsprung's disease and allied disorders, No. 1095 Jiefang Avenue, Qiaokou District, Wuhan, Hubei 430030, China
| |
Collapse
|
47
|
Ma X, Lu Y, Stoneking M, Xu S. Neanderthal adaptive introgression shaped LCT enhancer region diversity without linking to lactase persistence in East Asian populations. Proc Natl Acad Sci U S A 2025; 122:e2404393122. [PMID: 40063818 PMCID: PMC11929401 DOI: 10.1073/pnas.2404393122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 02/08/2025] [Indexed: 03/25/2025] Open
Abstract
Positive selection at the 2q21.3 enhancer region for lactase gene (LCT) expression in Europeans and Africans has long been attributed to selection for lactase persistence (LP), the capacity of adults to digest lactose in milk, presumably because of the benefits associated with milk consumption. While considered a classic example of gene-culture coevolution, recently doubts have been raised about the link between selection at 2q21.3 and LP. Analysis of additional populations could shed further light; here, we demonstrate that a haplotype spanning ~467 kb at the 2q21.3 locus has risen to high frequency in East Asians (~25%) but is absent from Africans and Europeans. This haplotype likely derived from Neanderthals and has been under positive selection in East Asians. The East Asian-specific haplotype is associated with alterations in LCT expression and promoter methylation in certain cell types, similar to what is observed with LP-associated haplotypes in Europeans. Moreover, its frequency is comparable to that of LP in East Asians, suggesting a potential association with LP in East Asians. However, it is highly unlikely that selection in East Asians was related to milk-drinking habits. We find that this haplotype impacts the expression of UBXN4, DARS1, and DARS1-AS1 in immune cells and is associated with neutrophil and white blood cell counts. Hence, the selection might be linked to certain aspects of immune function. This implies that selection on 2q21.3 has thus either occurred for different reasons in different populations or the selection observed in other populations is also not due to LP.
Collapse
Affiliation(s)
- Xixian Ma
- Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai200031, China
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen518055, China
| | - Yan Lu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai200032, China
| | - Mark Stoneking
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary Anthropology, LeipzigD04103, Germany
- Biométrie et Biologie Évolutive, Unité mixte de recherche 5558, CNRS & Université de Lyon, Lyon69622, France
| | - Shuhua Xu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Center for Evolutionary Biology, School of Life Sciences, Department of Liver Surgery and Transplantation Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai200032, China
- Ministry of Education Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai200032, China
| |
Collapse
|
48
|
de la Puente M, Casanova-Adán L, González-Bao J, Pardo-Seco J, Mosquera-Miguel A, Ambroa-Conde A, Ruiz-Ramírez J, Cabrejas-Olalla A, Boullón-Cassau M, Freire-Aradas A, Rodríguez A, Phillips C, Lareu MV. Evaluating the effect of marker panel sizes on estimation of bio-geographical co-ancestry proportions. Forensic Sci Int Genet 2025; 78:103275. [PMID: 40117914 DOI: 10.1016/j.fsigen.2025.103275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
A large number of ancestry-informative marker panels have been developed for forensic bio-geographical ancestry (BGA) analysis during the past decade, which offer valuable investigative tools for cold cases. The developed assays for capillary electrophoresis (CE) and massively parallel sequencing (MPS) focus on the differentiation of major populations, with MPS allowing larger numbers of markers that can be multiplexed at the same time and therefore improved differentiation of more closely related Eurasian populations. One limitation of BGA inference tools is the handling of co-ancestry in individuals with admixted backgrounds, which leads to two situations being indistinguishable: (i) the individual belongs to an admixed population, or (ii) the individual has recent ancestors from different populations. Accurate and precise co-ancestry estimates can help, as first or second-degree admixture would show a ∼ 50-50 % or ∼ 75-25 % ratio of co-ancestry proportions. Here we compared the co-ancestry proportion estimations obtained for the set of 2504 individuals from the 1000 Genomes Project with dedicated BGA and human identification (ID) assays of different sizes compared to those obtained with the > 500,000 SNP Affymetrix Human Origins panel as the point of reference for each individual. The results of the correlation analysis performed with > 500 admixed individuals indicate that panel size plays a major role in the accuracy of the co-ancestry estimates. Therefore, the large-scale forensic MPS ID panels we evaluated constitute a valuable alternative to small- and medium-scale BGA panels, especially when admixture is expected.
Collapse
Affiliation(s)
- M de la Puente
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain.
| | - L Casanova-Adán
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - J González-Bao
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - J Pardo-Seco
- Genetics, Vaccines and Pediatric Infectious Diseases Research Group (GENVIP) and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), Santiago de Compostela, Galicia, Spain; Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - A Mosquera-Miguel
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - A Ambroa-Conde
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - J Ruiz-Ramírez
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - A Cabrejas-Olalla
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - M Boullón-Cassau
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - A Freire-Aradas
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - A Rodríguez
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| | - C Phillips
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain; King's Forensics, Faculty of Life Sciences and Medicine, King's College, London, UK
| | - M V Lareu
- Forensic Genetics Unit, Institute of Forensic Sciences, Universidade de Santiago de Compostela, Spain
| |
Collapse
|
49
|
Topolska M, Beltran A, Lehner B. Deep indel mutagenesis reveals the impact of amino acid insertions and deletions on protein stability and function. Nat Commun 2025; 16:2617. [PMID: 40097423 PMCID: PMC11914627 DOI: 10.1038/s41467-025-57510-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 02/21/2025] [Indexed: 03/19/2025] Open
Abstract
Amino acid insertions and deletions (indels) are an abundant class of genetic variants. However, compared to substitutions, the effects of indels on protein stability are not well understood. To better understand indels here we analyse new and existing large-scale deep indel mutagenesis (DIM) of structurally diverse proteins. The effects of indels on protein stability vary extensively among and within proteins and are not well predicted by existing computational methods. To address this shortcoming we present INDELi, a series of models that combine experimental or predicted substitution effects and secondary structure information to provide good prediction of the effects of indels on both protein stability and pathogenicity. Moreover, quantifying the effects of indels on protein-protein interactions suggests that insertions can be an important class of gain-of-function variants. Our results provide an overview of the impact of indels on proteins and a method to predict their effects genome-wide.
Collapse
Affiliation(s)
- Magdalena Topolska
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- University Pompeu Fabra (UPF), Barcelona, Spain
| | - Antoni Beltran
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Ben Lehner
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- University Pompeu Fabra (UPF), Barcelona, Spain.
- Institució Catalana de Recerca i estudis Avançats (ICREA), Barcelona, Spain.
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
| |
Collapse
|
50
|
Clavell-Revelles P, Reese F, Carbonell-Sala S, Degalez F, Oliveros W, Arnan C, Guigó R, Melé M. Long-read transcriptomics of a diverse human cohort reveals widespread ancestry bias in gene annotation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643250. [PMID: 40166264 PMCID: PMC11956941 DOI: 10.1101/2025.03.14.643250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Accurate gene annotations are fundamental for interpreting genetic variation, cellular function, and disease mechanisms. However, current human gene annotations are largely derived from transcriptomic data of individuals with European ancestry, introducing potential biases that remain uncharacterized. Here, we generate over 800 million full-length reads with long-read RNA-seq in 43 lymphoblastoid cell line samples from eight genetically-diverse human populations and build a cross-ancestry gene annotation. We show that transcripts from non-European samples are underrepresented in reference gene annotations, leading to systematic biases in allele-specific transcript usage analyses. Furthermore, we show that personal genome assemblies enhance transcript discovery compared to the generic GRCh38 reference assembly, even though genomic regions unique to each individual are heavily depleted of genes. These findings underscore the urgent need for a more inclusive gene annotation framework that accurately represents global transcriptome diversity.
Collapse
Affiliation(s)
- Pau Clavell-Revelles
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Catalonia
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
- Universitat de Barcelona (UB), Barcelona, Catalonia
| | - Fairlie Reese
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Catalonia
| | - Sílvia Carbonell-Sala
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
| | - Fabien Degalez
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
| | - Winona Oliveros
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Catalonia
- Universitat de Barcelona (UB), Barcelona, Catalonia
| | - Carme Arnan
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
| | - Roderic Guigó
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Catalonia
- Universitat Pompeu Fabra (UPF), Barcelona, Catalonia
| | - Marta Melé
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Catalonia
| |
Collapse
|