1
|
Agrawal R, Al-Hiyari S, Hugh-White R, Hromas R, Patel Y, Williamson EA, Mootor MFE, Gonzalez A, Fu J, Haas R, Jordan M, Wickes BL, Mohammed G, Tian M, Doris MJ, Jobin C, Wernke KM, Pan Y, Yamaguchi TN, Herzon SB, Boutros PC, Liss MA. Colibactin Exerts Androgen-dependent and -independent Effects on Prostate Cancer. Eur Urol Oncol 2025; 8:716-730. [PMID: 39547899 PMCID: PMC12075626 DOI: 10.1016/j.euo.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/23/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND AND OBJECTIVE The etiology of prostate cancer (PC) is multifactorial and poorly understood. It has been suggested that colibactin-producing Escherichia coli positive for the pathogenicity island pks (pks+) initiate cancers via induction of genomic instability. In PC, androgens promote oncogenic translocations. Our aim was to investigate the association of pks+E. coli with PC diagnosis and molecular architecture, and its relationship with androgens. METHODS We quantified the association of pks+E. coli with PC diagnosis in a volunteer-sampled 235-person cohort from two institutional practices (UT San Antonio). We then used colibactin 742 and DNA/RNA sequencing to evaluate the effects of colibactin 742, dihydrotestosterone (DHT), and their combination in vitro. KEY FINDINGS AND LIMITATIONS Colibactin exposure was positively associated with PC diagnosis (p = 0.04) in our clinical cohort, and significantly increased replication fork stalling and fusions in vitro (p < 0.01). Combined in vitro exposure to colibactin 742 and DHT induced more somatic mutations of all types than exposure to either alone. The combination also elicited kataegis, with a higher density of somatic point mutations. Laboratory analyses were conducted using a single cell line, which limited our ability to fully recapitulate the complexity of PC etiology. CONCLUSIONS AND CLINICAL IMPLICATIONS Our findings are consistent with synergistic induction of genome instability and kataegis by colibactin 742 and DHT in cell culture. Colibactin-producing pks+ E. coli may plausibly contribute to PC etiology. PATIENT SUMMARY We investigated whether a bacterial toxin that is linked to colon cancer can also cause prostate cancer. Our results support this idea by showing a link between the toxin and prostate cancer diagnosis in a large patient population. We also found that this toxin causes genetic dysfunction in prostate cancer cells when combined with testosterone.
Collapse
Affiliation(s)
- Raag Agrawal
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA
| | - Sarah Al-Hiyari
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California-Los Angeles, Los Angeles, CA, USA
| | - Rupert Hugh-White
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California-Los Angeles, Los Angeles, CA, USA
| | - Robert Hromas
- Division of Hematology and Medical Oncology, Department of Medicine and the Mays Cancer Center, University of Texas Health-San Antonio, San Antonio, TX, USA
| | - Yash Patel
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California-Los Angeles, Los Angeles, CA, USA
| | - Elizabeth A Williamson
- Division of Hematology and Medical Oncology, Department of Medicine and the Mays Cancer Center, University of Texas Health-San Antonio, San Antonio, TX, USA
| | - Mohammed F E Mootor
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California-Los Angeles, Los Angeles, CA, USA
| | - Alfredo Gonzalez
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA
| | - Jianmin Fu
- Department of Microbiology and Immunology, University of Texas Health-San Antonio, San Antonio, TX, USA
| | - Roni Haas
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA
| | - Madison Jordan
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA
| | - Brian L Wickes
- Department of Microbiology, Immunology, and Molecular Genetics, University of Texas, San Antonia, TC, USA
| | - Ghouse Mohammed
- Office of Health Informatics and Analytics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Mao Tian
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA
| | - Molly J Doris
- Department of Urology, University of Texas Health-San Antonio, San Antonio, TX, USA
| | - Christian Jobin
- Departments of Medicine, Infectious Diseases and Immunology, and Anatomy and Cell Physiology, University of Florida, Gainesville, FL, USA
| | - Kevin M Wernke
- Department of Chemistry, Yale University, New Haven, CT, USA; Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Yu Pan
- Office of Health Informatics and Analytics, University of California-Los Angeles, Los Angeles, CA, USA
| | - Takafumi N Yamaguchi
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California-Los Angeles, Los Angeles, CA, USA
| | - Seth B Herzon
- Department of Chemistry, Yale University, New Haven, CT, USA; Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Paul C Boutros
- Department of Human Genetics, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California-Los Angeles, Los Angeles, CA, USA; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Michael A Liss
- Department of Urology, University of Texas Health-San Antonio, San Antonio, TX, USA.
| |
Collapse
|
2
|
d'Errico A, Vonk PJ, Wösten HAB, Lugones LG. Transposition of a non-autonomous element into the G β gene of Schizophyllum commune causes the streak mutation. Fungal Genet Biol 2025; 179:104007. [PMID: 40447071 DOI: 10.1016/j.fgb.2025.104007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/21/2025] [Accepted: 05/24/2025] [Indexed: 06/11/2025]
Abstract
Streak mutants of Schizophyllum commune are characterized by ropy, hyperbranching hyphae, suppressed aerial hyphae formation, and the production of pigments. Additionally, these mutants dikaryotize unilaterally, with the mutant fertilizing its compatible mating partner, but not accepting its nucleus. Here we show that a 512 bp non-autonomous transposable element had integrated in the Gβ protein of a streak mutant of S. commune. This element has the same 50 bp inverted repeat as an autonomous element, dubbed Bike transposon. Its transposase has homologues in various Agaricomycetes. Introducing the Gβ gene in the streak mutant restored the wild-type phenotype showing that the integration of the 512 bp element in the Gβ gene is responsible for the streak phenotype.
Collapse
Affiliation(s)
- Antonio d'Errico
- Microbiology, Department of Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands.
| | - Peter Jan Vonk
- Microbiology, Department of Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands.
| | - Han A B Wösten
- Microbiology, Department of Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands.
| | - Luis G Lugones
- Microbiology, Department of Biology, Utrecht University, Padualaan 8, 3584 CH, Utrecht, the Netherlands.
| |
Collapse
|
3
|
Wang F, Lu S, Xu W, Yang L. Deciphering the complex molecular architecture of the genetically modified soybean FG72 through paired-end whole genome sequencing. FOOD CHEMISTRY. MOLECULAR SCIENCES 2025; 10:100238. [PMID: 39839759 PMCID: PMC11750281 DOI: 10.1016/j.fochms.2024.100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/23/2025]
Abstract
The clear molecular characterization of genetically modified (GM) plants and animals is a prerequisite for obtaining regulatory approval and safety certification for commercial cultivation. This characterization includes the identification of the transferred DNA (T-DNA) insertion site, its flanking sequences, the copy number of inserted genes, and the detection of any unintended genomic alterations accompanying the transformation process. In this study, we performed a comprehensive molecular characterization of the well-known GM soybean event FG72 using paired-end whole-genome sequencing (PE-WGS). We examined the T-DNA insertion site, flanking sequences, the entire structure and copy number of the T-DNA integration, the presence of plasmid backbone sequences, and genome-wide structural variations (SVs). Our analysis revealed that the T-DNA integrated into two distinct sites on chromosome 15 of the host genome, accompanied by a translocation of host genomic sequences. One site harbored a partial T-DNA integration, while the other site contained two tandem repeats of the full T-DNA. Importantly, no plasmid backbone sequences were detected in the host genome, indicating a clean T-DNA integration during the biolistic transformation process. Furthermore, we identified numerous genome-wide SVs, with chromosome 15 ranking second among all 20 chromosomes in terms of SV frequency, and most of these variations occurring within gene-coding regions. These results provide a refined and comprehensive molecular characterization of the FG72 soybean event, which could further support its commercial approval and cultivation. Our work highlights the utility of the PE-WGS approach as a sensitive and labor-efficient alternative to conventional molecular characterization techniques, generating comprehensive data to facilitate the safety assessment of GM crops during research and commercialization pipelines.
Collapse
Affiliation(s)
- Fan Wang
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Shengtao Lu
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China
- Zhejiang Yuzhi Biotechnology Company Ltd, Ningbo 315032, PR China
| | - Wenting Xu
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Litao Yang
- Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, PR China
- Zhejiang Yuzhi Biotechnology Company Ltd, Ningbo 315032, PR China
- Yazhou Bay Institute of Deepsea Sci-Tech, Shanghai Jiao Tong University, Sanya 572024, PR China
| |
Collapse
|
4
|
Wang D, Zhang T, Zhang M, Chen Q, Yan M, Ma S, Wang J, Zhang X, Ma K, Xu L, Huang X. Identification of Genomic Structural Variations in Xinjiang Brown Cattle by Deep Sequencing and Their Association with Body Conformation Traits. Int J Mol Sci 2025; 26:5234. [PMID: 40508044 PMCID: PMC12154528 DOI: 10.3390/ijms26115234] [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: 03/19/2025] [Revised: 05/26/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
Xinjiang Brown cattle is an elite dual-purpose breed (raised for dairy and beef) developed in China. To elucidate its genomic architecture, we conducted whole-genome resequencing of 169 Xinjiang Brown cattle, followed by structural variation (SV) detection and a genome-wide association study (GWAS). We identified 71,668 SVs, among which deletions were the most prevalent, followed by translocations, inversions, duplications, and insertions. We further identified 1286 high-frequency SVs involving 2016 protein-coding genes. Through functional enrichment analysis of these genes, we revealed associations of genetic variation at genomic positions near genes implicated in immune response and disease resistance (NFKBIZ and PTPRT), growth and development (HDAC4 and MEF2A), and milk production (TP63, FABP4, and MEF2A). GWAS analysis of 31 body conformation traits revealed 58 SVs significantly associated with five traits (chest width, rear udder width, udder depth, rump width, and heel depth) at the genome-wide level. Additionally, nine candidate genes (CLINT1, EBF1, PAM16, GRIP1, CFAP54, SLC22A16, DOK5, ETAA1, and IPMK) were identified as potentially involved in the genetic regulation of body conformation traits. These findings provide novel insights for genetic improvement strategies and indicate that precision breeding could further enhance the production performance of this breed in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Lei Xu
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (D.W.); (T.Z.); (M.Z.); (Q.C.); (M.Y.); (S.M.); (J.W.); (X.Z.); (K.M.)
| | - Xixia Huang
- College of Animal Science, Xinjiang Agricultural University, Urumqi 830052, China; (D.W.); (T.Z.); (M.Z.); (Q.C.); (M.Y.); (S.M.); (J.W.); (X.Z.); (K.M.)
| |
Collapse
|
5
|
Kim JP, Cho M, Kim C, Lee H, Jang B, Jung SH, Kim Y, Koh IG, Kim S, Shin D, Lee EH, Lee JY, Park Y, Jang H, Kim BH, Ham H, Kim B, Kim Y, Cho AH, Raj T, Kim HJ, Na DL, Seo SW, An JY, Won HH. Whole-genome sequencing analyses suggest novel genetic factors associated with Alzheimer's disease and a cumulative effects model for risk liability. Nat Commun 2025; 16:4870. [PMID: 40419521 PMCID: PMC12106753 DOI: 10.1038/s41467-025-59949-y] [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: 05/26/2024] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
Genome-wide association studies (GWAS) on Alzheimer's disease (AD) have predominantly focused on identifying common variants in Europeans. Here, we performed whole-genome sequencing (WGS) of 1,559 individuals from a Korean AD cohort to identify various genetic variants and biomarkers associated with AD. Our GWAS analysis identified a previously unreported locus for common variants (APCDD1) associated with AD. Our WGS analysis was extended to explore the less-characterized genetic factors contributing to AD risk. We identified rare noncoding variants located in cis-regulatory elements specific to excitatory neurons associated with cognitive impairment. Moreover, structural variation analysis showed that short tandem repeat expansion was associated with an increased risk of AD, and copy number variant at the HPSE2 locus showed borderline statistical significance. APOE ε4 carriers with high polygenic burden or structural variants exhibited severe cognitive impairment and increased amyloid beta levels, suggesting a cumulative effects model of AD risk.
Collapse
Affiliation(s)
- Jun Pyo Kim
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Minyoung Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Chanhee Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - Hyunwoo Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Beomjin Jang
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sang-Hyuk Jung
- Department of Medical Informatics, Kangwon National University College of Medicine, Chuncheon, Republic of Korea
| | - Yujin Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - In Gyeong Koh
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - Seoyeon Kim
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea
| | - Daeun Shin
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Eun Hye Lee
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Alzheimer Disease Research Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - YoungChan Park
- Division of Bio Bigdata, Department of Precision Medicine, Korea National Institution of Health, Cheongju, Republic of Korea
| | - Hyemin Jang
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Seoul National University Hospital, Seoul National University School of Medicine, Seoul, Republic of Korea
| | - Bo-Hyun Kim
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Hongki Ham
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Beomsu Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Yujin Kim
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - A-Hyun Cho
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea
| | - Towfique Raj
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Nash Family Department of Neuroscience & Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Estelle and Daniel Maggin Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hee Jin Kim
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Duk L Na
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea
| | - Sang Won Seo
- Alzheimer's Disease Convergence Research Center, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
- Neuroscience Center, Samsung Medical Center, Seoul, Republic of Korea.
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea.
| | - Joon-Yong An
- Department of Integrated Biomedical and Life Science, Korea University, Seoul, Republic of Korea.
- L-HOPE Program for Community-Based Total Learning Health Systems, Korea University, Seoul, Republic of Korea.
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea.
| | - Hong-Hee Won
- Department of Digital Health, Samsung Advanced Institute for Health Sciences & Technology (SAIHST), Sungkyunkwan University, Seoul, Republic of Korea.
- Samsung Genome Institute, Samsung Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
6
|
Song GY, Park JH, Kang SR, Han SJ, Jung Y, Son M, Jang HC, Kim M, Ahn SY, Jung SH, Ahn JS, Lee JJ, Kim HJ, Yang DH. Clinical Implication of Sequential Circulating Tumor DNA Assessments for the Treatment of Diffuse Large B-Cell Lymphoma. Cancers (Basel) 2025; 17:1734. [PMID: 40507216 PMCID: PMC12153856 DOI: 10.3390/cancers17111734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2025] [Revised: 05/09/2025] [Accepted: 05/15/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES Circulating tumor DNA (ctDNA) has emerged as a promising biomarker for non-invasive tumor monitoring in diffuse large B-cell lymphoma (DLBCL). METHODS In this study, 52 patients with newly diagnosed advanced-stage DLBCL treated with R-CHOP underwent serial ctDNA analysis at baseline, interim (after three cycles), and end of treatment. The prognostic significance of ctDNA dynamics was evaluated, and its predictive value was compared with the PET/CT response. RESULTS Targeted next-generation sequencing revealed baseline ctDNA in 98.1% of patients, with 74.7% concordance to tumor tissue genotyping. Higher baseline ctDNA levels correlated with elevated LDH, older age, and high IPI scores. A ≥2-log reduction in ctDNA at interim was significantly associated with improved overall survival (p = 0.004), though not with progression-free survival. Notably, combining interim ctDNA dynamics with PET/CT results enhanced the predictive accuracy for treatment outcomes, particularly among patients with partial metabolic responses. CONCLUSIONS These findings support the clinical utility of ctDNA for dynamic risk assessment in DLBCL, and suggest that integrating ctDNA with imaging biomarkers may guide more personalized therapeutic strategies. Further validation using highly sensitive ctDNA assays is warranted to optimize its role in routine clinical practice.
Collapse
Affiliation(s)
- Ga-Young Song
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea; (G.-Y.S.); (H.C.J.); (M.K.); (S.-Y.A.); (S.-H.J.); (J.-S.A.); (J.-J.L.); (H.-J.K.)
| | - Joo Heon Park
- Department of Laboratory Medicine, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea
| | - Sae-Ryung Kang
- Department of Nuclear Medicine, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea;
| | - Seung Jung Han
- Dxome Co. Ltd., Bundang-gu, Seongnam-si 13558, Gyeonggi-do, Republic of Korea; (S.J.H.); (Y.J.); (M.S.)
| | - Youjin Jung
- Dxome Co. Ltd., Bundang-gu, Seongnam-si 13558, Gyeonggi-do, Republic of Korea; (S.J.H.); (Y.J.); (M.S.)
| | - Minuk Son
- Dxome Co. Ltd., Bundang-gu, Seongnam-si 13558, Gyeonggi-do, Republic of Korea; (S.J.H.); (Y.J.); (M.S.)
| | - Ho Cheol Jang
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea; (G.-Y.S.); (H.C.J.); (M.K.); (S.-Y.A.); (S.-H.J.); (J.-S.A.); (J.-J.L.); (H.-J.K.)
| | - Mihee Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea; (G.-Y.S.); (H.C.J.); (M.K.); (S.-Y.A.); (S.-H.J.); (J.-S.A.); (J.-J.L.); (H.-J.K.)
| | - Seo-Yeon Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea; (G.-Y.S.); (H.C.J.); (M.K.); (S.-Y.A.); (S.-H.J.); (J.-S.A.); (J.-J.L.); (H.-J.K.)
| | - Sung-Hoon Jung
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea; (G.-Y.S.); (H.C.J.); (M.K.); (S.-Y.A.); (S.-H.J.); (J.-S.A.); (J.-J.L.); (H.-J.K.)
| | - Jae-Sook Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea; (G.-Y.S.); (H.C.J.); (M.K.); (S.-Y.A.); (S.-H.J.); (J.-S.A.); (J.-J.L.); (H.-J.K.)
| | - Je-Jung Lee
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea; (G.-Y.S.); (H.C.J.); (M.K.); (S.-Y.A.); (S.-H.J.); (J.-S.A.); (J.-J.L.); (H.-J.K.)
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea; (G.-Y.S.); (H.C.J.); (M.K.); (S.-Y.A.); (S.-H.J.); (J.-S.A.); (J.-J.L.); (H.-J.K.)
| | - Deok-Hwan Yang
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Medical School of Chonnam National University, Hwasun-gun 58128, Jeollanam-do, Republic of Korea; (G.-Y.S.); (H.C.J.); (M.K.); (S.-Y.A.); (S.-H.J.); (J.-S.A.); (J.-J.L.); (H.-J.K.)
| |
Collapse
|
7
|
Boulling A, Corbeau J, Grohs C, Barbat A, Mortier J, Taussat S, Plassard V, Leclerc H, Fritz S, Leymarie C, Bourgeois-Brunel L, Ducos A, Guatteo R, Boichard D, Boussaha M, Capitan A. A bovine model of rhizomelic chondrodysplasia punctata caused by a deep intronic splicing variant in the GNPAT gene. Genet Sel Evol 2025; 57:23. [PMID: 40394457 PMCID: PMC12090490 DOI: 10.1186/s12711-025-00969-z] [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: 06/26/2024] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Genetic defects that occur naturally in livestock species provide valuable models for investigating the molecular mechanisms underlying rare human diseases. Livestock breeds are subject to the regular emergence of recessive genetic defects due to genetic drift and recent inbreeding. At the same time, their large population sizes provide easy access to case and control individuals and to massive amounts of pedigree, genomic and phenotypic information recorded for management and selection purposes. In this study, we investigated a lethal form of recessive chondrodysplasia observed in 21 stillborn calves of the Aubrac beef cattle breed. RESULTS Detailed examinations of three affected calves revealed proximal limb shortening, epiphyseal calcific deposits, and other pathological signs consistent with human rhizomelic chondrodysplasia punctata, a rare peroxisomal disorder caused by recessive variants in one of five genes (AGPS, FAR1, GNPAT, PEX5, and PEX7). Using homozygosity mapping, whole genome sequencing of two affected individuals, and filtering for variants found in 1867 control genomes, we reduced the list of candidate variants to a single deep intronic substitution in GNPAT (NC_037355.1:g.4039268G > A on chromosome 28 of the ARS-UCD1.2 bovine genome assembly). For verification, we performed large-scale genotyping of this variant using a custom SNP array and found a perfect genotype-phenotype correlation in 21 cases and 26 of their parents, and a complete absence of homozygotes in 1195 unaffected Aubrac controls. The g.4039268A allele segregated at a frequency of 2.6% in this population and was absent in 375,535 additional individuals from 17 breeds. Then, using in vivo and in vitro analyses, we demonstrated that the derived allele activates cryptic splice sites within intron 11 resulting in abnormal transcripts. Finally, by mining the wealth of records available in the French bovine database, we also reported suggestive effects on juvenile mortality (and not just stillbirth) in homozygotes and on muscle development in heterozygotes, which merit further investigation. CONCLUSIONS We report the first spontaneous large animal model of rhizomelic chondrodysplasia punctata and provide a diagnostic test to select against this defect in cattle. Our work also brings interesting insights into the molecular consequences of complete or partial GNPAT insufficiency in mammals.
Collapse
Affiliation(s)
- Arnaud Boulling
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.
| | - Julien Corbeau
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- Oniris, INRAE, BIOEPAR, 44300, Nantes, France
| | - Cécile Grohs
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Anne Barbat
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Jérémy Mortier
- Service d'imagerie Médicale, DEPEC, Ecole Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Sébastien Taussat
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- ELIANCE, 75012, Paris, France
| | - Vincent Plassard
- Service d'imagerie Médicale, DEPEC, Ecole Nationale Vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Hélène Leclerc
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- ELIANCE, 75012, Paris, France
| | - Sébastien Fritz
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
- ELIANCE, 75012, Paris, France
| | | | | | - Alain Ducos
- GenPhySE, Université de Toulouse, INRAE, ENVT, 31326, Castanet Tolosan, France
| | | | - Didier Boichard
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Mekki Boussaha
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Aurélien Capitan
- Université Paris Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.
- ELIANCE, 75012, Paris, France.
| |
Collapse
|
8
|
Zhou Y, Anthony R, Wang S, Xia H, Ou X, Zhao B, Song Y, Zheng Y, He P, Liu D, Zhao Y, van Soolingen D. Understanding the epidemiology and pathogenesis of Mycobacterium tuberculosis with non-redundant pangenome of epidemic strains in China. PLoS One 2025; 20:e0324152. [PMID: 40388514 DOI: 10.1371/journal.pone.0324152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 04/21/2025] [Indexed: 05/21/2025] Open
Abstract
Tuberculosis is a major public health threat resulting in more than one million lives lost every year. Many challenges exist to defeat this deadly infectious disease which address the importance of a thorough understanding of the biology of the causative agent Mycobacterium tuberculosis (MTB). We generated a non-redundant pangenome of 420 epidemic MTB strains from China including 344 Lineage 2 strains, 69 Lineage 4 strains, six Lineage 3 strains, and one Lineage 1 strain. We estimate that MTB strains have a pangenome of 4,278 genes encoding 4,183 proteins, of which 3,438 are core genes. However, due to 99,694 interruptions in 2,447 coding genes, we can only confidently confirm 1,651 of these genes are translated in all samples. Of these interruptions, 67,315 (67.52%) could be classified by various genetic variations detected by currently available tools, and more than half of them are due to structural variations, mostly small indels. Assuming a proportion of these interruptions are artifacts, the number of active core genes would still be much lower than 3,438. We further described differential evolutionary patterns of genes under the influences of selective pressure, population structure and purifying selection. While selective pressure is ubiquitous among these coding genes, evolutionary adaptations are concentrated in 1,310 genes. Genes involved in cell wall biogenesis are under the strongest selective pressure, while the biological process of disruption of host organelles indicates the direction of the most intensive positive selection. This study provides a comprehensive view on the genetic diversity and evolutionary patterns of coding genes in MTB which may deepen our understanding of its epidemiology and pathogenicity.
Collapse
Affiliation(s)
- Yang Zhou
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
- Radboudumc Research Institute, Radboud University, Houtlaan XZ, Nijmegen, The Netherlands
| | - Richard Anthony
- National Tuberculosis Reference Laboratory, Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Shengfen Wang
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
| | - Hui Xia
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
| | - Xichao Ou
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
| | - Bing Zhao
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
| | - Yuanyuan Song
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
| | - Yang Zheng
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
| | - Ping He
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
| | - Dongxin Liu
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
| | - Yanlin Zhao
- National Center for Tuberculosis Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing, China
| | - Dick van Soolingen
- Radboudumc Research Institute, Radboud University, Houtlaan XZ, Nijmegen, The Netherlands
| |
Collapse
|
9
|
Wu B, Luo D, Yue Y, Yan H, He M, Ma X, Zhao B, Xu B, Zhu J, Wang J, Jia J, Sun M, Xie Z, Wang X, Huang L. New insights into the cold tolerance of upland switchgrass by integrating a haplotype-resolved genome and multi-omics analysis. Genome Biol 2025; 26:128. [PMID: 40369670 PMCID: PMC12076936 DOI: 10.1186/s13059-025-03604-8] [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: 09/25/2024] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Switchgrass (Panicum virgatum L.) is a bioenergy and forage crop. Upland switchgrass exhibits superior cold tolerance compared to the lowland ecotype, but the underlying molecular mechanisms remain unclear. RESULTS Here, we present a high-quality haplotype-resolved genome of the upland ecotype "Jingji31." We then conduct multi-omics analysis to explore the mechanism underlying its cold tolerance. By comparative transcriptome analysis of the upland and lowland ecotypes, we identify many genes with ecotype-specific differential expression, particularly members of the cold-responsive (COR) gene family, under cold stress. Notably, AFB1, ATL80, HOS10, and STRS2 gene families show opposite expression changes between the two ecotypes. Based on the haplotype-resolved genome of "Jingji31," we detect more cold-induced allele-specific expression genes in the upland ecotype than in the lowland ecotype, and these genes are significantly enriched in the COR gene family. By genome-wide association study, we detect an association signal related to the overwintering rate, which overlaps with a selective sweep region containing a cytochrome P450 gene highly expressed under cold stress. Heterologous overexpression of this gene in rice alleviates leaf chlorosis and wilting under cold stress. We also verify that expression of this gene is suppressed by a structural variation in the promoter region. CONCLUSIONS Based on the high-quality haplotype-resolved genome and multi-omics analysis of upland switchgrass, we characterize candidate genes responsible for cold tolerance. This study advances our understanding of plant cold tolerance, which provides crop breeding for improved cold tolerance.
Collapse
Affiliation(s)
- Bingchao Wu
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Dan Luo
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yuesen Yue
- Institute of Grassland, Flower and Ecology, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China
| | - Haidong Yan
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Min He
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xixi Ma
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Bingyu Zhao
- College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Bin Xu
- College of Grassland Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jie Zhu
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Jing Wang
- Key Laboratory for Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610061, China
| | - Jiyuan Jia
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Min Sun
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
- Institute of Advanced Study, Chengdu University, Chengdu, 610106, China
| | - Zheni Xie
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xiaoshan Wang
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Linkai Huang
- College of Grassland Science and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
10
|
Acford-Palmer H, Tadesse FG, Manko E, Phelan JE, Higgins M, Osborne A, Kristan M, Walker T, Bousema T, Messenger LA, Clark TG, Campino S. Genome wide population genetics and molecular surveillance of insecticide resistance in Anopheles stephensi mosquitoes from Awash Sebat Kilo in Ethiopia. Sci Rep 2025; 15:16443. [PMID: 40355632 PMCID: PMC12069653 DOI: 10.1038/s41598-025-95814-0] [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: 03/19/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
Since the detection of the Asian mosquito Anopheles stephensi in Dijbouti in 2012, it has spread throughout the Horn of Africa. This invasive vector continues to expand across the continent and is a significant threat to malaria control programs. Vector control methods, including insecticide-treated nets and indoor residual spraying, have substantially reduced the malaria burden. However, the increasing prevalence of mosquitoes resistant to insecticides, including An. stephensi populations, undermines ongoing malaria elimination efforts. Understanding population structure, gene flow between populations, and the distribution of insecticide resistance mutations is essential for guiding effective malaria control strategies. Here, we generated whole genome sequencing data for An. stephensi sourced from Awash Sebat Kilo, Ethiopia (n = 27) and compared with South Asian populations (n = 45; India and Pakistan) to assess genomic diversity, population structure, and uncovering insecticide resistance mutations. Population structure analysis using genome-wide single nucleotide polymorphisms (n = 15,533,476) revealed Ethiopian isolates clustering as a distinct ancestral group, separate from South Asian isolates. Three insecticide resistance-associated SNPs (gaba gene: A296S and V327I; vgsc L1014F) were detected. Evidence of ongoing selection was found in several loci, including genes previously associated with neonicotinoids, ivermectin, DDT, and pyrethroid resistance. This study represents the first whole genome population genetics study of invasive An. stephensi, revealing genomic differences from South Asian populations, which can be used for future assessments of vector population dispersal and detection of insecticide resistance mechanisms.
Collapse
Affiliation(s)
- Holly Acford-Palmer
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Fitsum G Tadesse
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Malaria and NTD Directorate, Armauer Hansen Research Institute, ALERT Hospital Compound, Addis Ababa, Ethiopia
| | - Emilia Manko
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Jody E Phelan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Matthew Higgins
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Ashley Osborne
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Mojca Kristan
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
| | - Thomas Walker
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, CV4 7AL, UK
| | - Teun Bousema
- Department of Medical Microbiology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Louisa A Messenger
- Department of Environmental and Occupational Health, School of Public Health, University of Nevada, Las Vegas, Las Vegas, USA
- Parasitology and Vector Biology (PARAVEC), School of Public Health, University of Nevada, Las Vegas, NV, USA
| | - Taane G Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, UK
| | - Susana Campino
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, UK.
| |
Collapse
|
11
|
Skelly DA, Graham JP, Cheng M, Furuta M, Walter A, Stoklasek TA, Yang H, Stearns TM, Poirion O, Zhang JG, Grassmann JDS, Luo D, Flynn WF, Courtois ET, Chang CH, Serreze DV, Menghi F, Reinholdt LG, Liu ET. Mapping the genetic landscape establishing a tumor immune microenvironment favorable for anti-PD-1 response. Cell Rep 2025; 44:115698. [PMID: 40343794 DOI: 10.1016/j.celrep.2025.115698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/03/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
Identifying host genetic factors modulating immune checkpoint inhibitor (ICI) efficacy is experimentally challenging. Our approach, utilizing the Collaborative Cross mouse genetic resource, fixes the tumor genomic configuration while varying host genetics. We find that response to anti-PD-1 (aPD1) immunotherapy is significantly heritable in four distinct murine tumor models (H2: 0.18-0.40). For the MC38 colorectal carcinoma system, we map four significant ICI response quantitative trait loci (QTLs) with significant epistatic interactions. The differentially expressed genes within these QTLs that define responder genetics are highly enriched for processes involving antigen processing and presentation, allograft rejection, and graft vs. host disease (all p < 1 × 10-10). Functional blockade of two top candidate immune targets, GM-CSF and IL-2RB, completely abrogates the MC38 transcriptional response to aPD1 therapy. Thus, our in vivo experimental platform is a powerful approach for discovery of host genetic factors that establish the tumor immune microenvironment propitious for ICI response.
Collapse
Affiliation(s)
- Daniel A Skelly
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - John P Graham
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | - Mayuko Furuta
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Andrew Walter
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | | | | | - Timothy M Stearns
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Olivier Poirion
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Ji-Gang Zhang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Jessica D S Grassmann
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Diane Luo
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - William F Flynn
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Elise T Courtois
- Single Cell Biology Lab, The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; OB/Gyn Department, UConn Health, Farmington, CT 06032, USA
| | - Chih-Hao Chang
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - David V Serreze
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Francesca Menghi
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Laura G Reinholdt
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME 04609, USA
| | - Edison T Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA.
| |
Collapse
|
12
|
Kinnersley B, Jung J, Cornish AJ, Chubb D, Laxton R, Frangou A, Gruber AJ, Sud A, Caravagna G, Sottoriva A, Wedge DC, Booth T, Al-Sarraj S, Lawrence SED, Albanese E, Anichini G, Baxter D, Boukas A, Chowdhury YA, D'Urso P, Corns R, Dapaah A, Edlmann E, Greenway F, Grundy P, Hill CS, Jenkinson MD, Trichinopoly Krishna S, Smith S, Manivannan S, Martin AJ, Matloob S, Mukherjee S, O'Neill K, Plaha P, Pollock J, Price S, Rominiyi O, Sachdev B, Saeed F, Sinha S, Thorne L, Ughratdar I, Whitfield P, Youshani AS, Bulbeck H, Arumugam P, Houlston R, Ashkan K. Genomic landscape of diffuse glioma revealed by whole genome sequencing. Nat Commun 2025; 16:4233. [PMID: 40335506 PMCID: PMC12059081 DOI: 10.1038/s41467-025-59156-9] [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: 10/27/2023] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
Diffuse gliomas are the commonest malignant primary brain tumour in adults. Herein, we present analysis of the genomic landscape of adult glioma, by whole genome sequencing of 403 tumours (256 glioblastoma, 89 astrocytoma, 58 oligodendroglioma; 338 primary, 65 recurrence). We identify an extended catalogue of recurrent coding and non-coding genetic mutations that represents a source for future studies and provides a high-resolution map of structural variants, copy number changes and global genome features including telomere length, mutational signatures and extrachromosomal DNA. Finally, we relate these to clinical outcome. As well as identifying drug targets for treatment of glioma our findings offer the prospect of improving treatment allocation with established targeted therapies.
Collapse
Affiliation(s)
- Ben Kinnersley
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK.
- UCL Cancer Institute, 72 Huntley St, WC1E 6DD, London, UK.
| | - Josephine Jung
- Institute of Psychiatry, Psychology and Neurosciences, Kings College London, Strand, WC2R 2LS, London, UK.
- Department of Neurosurgery, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK.
| | - Alex J Cornish
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Daniel Chubb
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Ross Laxton
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK
| | - Anna Frangou
- Cancer Genomics, Big Data Institute, Nuffield Department of Medicine, Old Road Campus, OX3 7LF, Oxford, UK
| | - Andreas J Gruber
- Department of Biology, University of Konstanz, Konstanz, 78464, Germany
| | - Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Giulio Caravagna
- Evolutionary Genomics and Modelling Lab, Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Andrea Sottoriva
- Evolutionary Genomics and Modelling Lab, Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - David C Wedge
- Manchester Cancer Research Centre, University of Manchester, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Thomas Booth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas's Hospital, London, UK
- Department of Neuroradiology, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK
| | - Safa Al-Sarraj
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK
| | - Samuel E D Lawrence
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Erminia Albanese
- Department of Neurosurgery, Royal Stoke University Hospital, Newcastle Road, ST4 6QG, Stoke-on-Trent, UK
| | - Giulio Anichini
- Imperial College Healthcare NHS Trust, Charing Cross Hospital, 3S corridor, Fulham Palace Road, London, W6 8RF, UK
| | - David Baxter
- Department of Neurosurgery, Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, HA7 4LP, UK
| | - Alexandros Boukas
- Department of Neurosurgery, John Radcliffe Hospital, Headley Way, Headington, OX3 9DU, Oxford, UK
| | - Yasir A Chowdhury
- Department of Neurosurgery, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, B15 2GW, Birmingham, UK
| | - Pietro D'Urso
- Department of Neurosurgery, Manchester Royal Infirmary, Oxford Rd, M13 9WL, Manchester, UK
| | - Robert Corns
- Department of Neurosurgery, Leeds General Infirmary, Great George St, LS1 3EX, Leeds, UK
| | - Andrew Dapaah
- Department of Neurosurgery, Queen's Medical Centre NHS Trust, Derby Road, Lenton, NG7 2UH, Nottingham, UK
| | - Ellie Edlmann
- South West Neurosurgery Unit, University Hospitals Plymouth NHS Trust, Derriford Road, Crownhill, PL6 8DH, Plymouth, UK
| | - Fay Greenway
- Department of Neurosurgery, St. George's University Hospitals NHS Foundation Trust, Blackshaw Rd, SW17 0QT, London, UK
| | - Paul Grundy
- Department of Neurosurgery, Southampton General Hospital, Tremona Road, SO16 6YD, Southampton, UK
| | - Ciaran S Hill
- UCL Cancer Institute, 72 Huntley St, WC1E 6DD, London, UK
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, WC1N 3BG, London, UK
| | - Michael D Jenkinson
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Lower Lane, Fazakerley, L9 7LJ, Liverpool, UK
| | - Sandhya Trichinopoly Krishna
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Lower Lane, Fazakerley, L9 7LJ, Liverpool, UK
| | - Stuart Smith
- Department of Neurosurgery, Queen's Medical Centre NHS Trust, Derby Road, Lenton, NG7 2UH, Nottingham, UK
| | - Susruta Manivannan
- Department of Neurosurgery, Southampton General Hospital, Tremona Road, SO16 6YD, Southampton, UK
| | - Andrew J Martin
- Department of Neurosurgery, St. George's University Hospitals NHS Foundation Trust, Blackshaw Rd, SW17 0QT, London, UK
| | - Samir Matloob
- Department of Neurosurgery, Queen's Hospital Romford, Rom Valley Way, RM7 0AG, Romford, UK
| | - Soumya Mukherjee
- Department of Neurosurgery, Addenbrookes Hospital, Hills Rd, CB2 0QQ, Cambridge, UK
| | - Kevin O'Neill
- Imperial College Healthcare NHS Trust, Charing Cross Hospital, 3S corridor, Fulham Palace Road, London, W6 8RF, UK
| | - Puneet Plaha
- Department of Neurosurgery, John Radcliffe Hospital, Headley Way, Headington, OX3 9DU, Oxford, UK
| | - Jonathan Pollock
- Department of Neurosurgery, Queen's Hospital Romford, Rom Valley Way, RM7 0AG, Romford, UK
| | - Stephen Price
- Department of Neurosurgery, Addenbrookes Hospital, Hills Rd, CB2 0QQ, Cambridge, UK
| | - Ola Rominiyi
- Department of Neurosurgery, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Rd, Broomhall, S10 2JF, Sheffield, UK
| | - Bobby Sachdev
- Department of Neurosurgery, Royal Stoke University Hospital, Newcastle Road, ST4 6QG, Stoke-on-Trent, UK
| | - Fozia Saeed
- Department of Neurosurgery, Leeds General Infirmary, Great George St, LS1 3EX, Leeds, UK
| | - Saurabh Sinha
- Department of Neurosurgery, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Rd, Broomhall, S10 2JF, Sheffield, UK
| | - Lewis Thorne
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, WC1N 3BG, London, UK
| | - Ismail Ughratdar
- Department of Neurosurgery, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, B15 2GW, Birmingham, UK
| | - Peter Whitfield
- South West Neurosurgery Unit, University Hospitals Plymouth NHS Trust, Derriford Road, Crownhill, PL6 8DH, Plymouth, UK
| | - Amir Saam Youshani
- Department of Neurosurgery, Manchester Royal Infirmary, Oxford Rd, M13 9WL, Manchester, UK
| | - Helen Bulbeck
- Brainstrust, 4 Yvery Court, Castle Road, PO31 7QG, Cowes, Isle of Wight, UK
| | | | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Keyoumars Ashkan
- Institute of Psychiatry, Psychology and Neurosciences, Kings College London, Strand, WC2R 2LS, London, UK.
- Department of Neurosurgery, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK.
| |
Collapse
|
13
|
Silaiyiman S, Liu J, Wu J, Ouyang L, Cao Z, Shen C. A Systematic Review of the Advances and New Insights into Copy Number Variations in Plant Genomes. PLANTS (BASEL, SWITZERLAND) 2025; 14:1399. [PMID: 40364428 PMCID: PMC12073271 DOI: 10.3390/plants14091399] [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: 03/05/2025] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025]
Abstract
Copy number variations (CNVs), as an important structural variant in genomes, are widely present in plants, affecting their phenotype and adaptability. In recent years, CNV research has not only focused on changes in gene copy numbers but has also been linked to complex mechanisms such as genome rearrangements, transposon activity, and environmental adaptation. The advancement in sequencing technologies has made the detection and analysis of CNVs more efficient, not only revealing their crucial roles in plant disease resistance, adaptability, and growth development, but also demonstrating broad application potential in crop improvement, particularly in selective breeding and genomic selection. By studying CNV changes during the domestication process, researchers have gradually recognized the important role of CNVs in plant domestication and evolution. This article reviews the formation mechanisms of CNVs in plants, methods for their detection, their relationship with plant traits, and their applications in crop improvement. It emphasizes future research directions involving the integration of multi-omics to provide new perspectives on the structure and function of plant genomes.
Collapse
Affiliation(s)
- Saimire Silaiyiman
- Guangdong Provincial Key Laboratory for Green Agricultural Production and Intelligent Equipment, College of Biological and Food Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China; (S.S.); (J.L.); (J.W.); (L.O.)
- College of Life and Geographic Sciences, Kashi University, Kashi 844000, China
- Key Laboratory of Biological Resources and Ecology of Pamirs Plateau in Xinjiang Uygur Autonomous Region, Kashi 844000, China
| | - Jiaxuan Liu
- Guangdong Provincial Key Laboratory for Green Agricultural Production and Intelligent Equipment, College of Biological and Food Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China; (S.S.); (J.L.); (J.W.); (L.O.)
- College of Life and Geographic Sciences, Kashi University, Kashi 844000, China
- Key Laboratory of Biological Resources and Ecology of Pamirs Plateau in Xinjiang Uygur Autonomous Region, Kashi 844000, China
| | - Jiaxin Wu
- Guangdong Provincial Key Laboratory for Green Agricultural Production and Intelligent Equipment, College of Biological and Food Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China; (S.S.); (J.L.); (J.W.); (L.O.)
- College of Life and Geographic Sciences, Kashi University, Kashi 844000, China
- Key Laboratory of Biological Resources and Ecology of Pamirs Plateau in Xinjiang Uygur Autonomous Region, Kashi 844000, China
| | - Lejun Ouyang
- Guangdong Provincial Key Laboratory for Green Agricultural Production and Intelligent Equipment, College of Biological and Food Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China; (S.S.); (J.L.); (J.W.); (L.O.)
- College of Life and Geographic Sciences, Kashi University, Kashi 844000, China
| | - Zheng Cao
- Maoming Agricultural Science and Technology Extension Center, Maoming 525000, China;
| | - Chao Shen
- Guangdong Provincial Key Laboratory for Green Agricultural Production and Intelligent Equipment, College of Biological and Food Engineering, Guangdong University of Petrochemical Technology, Maoming 525000, China; (S.S.); (J.L.); (J.W.); (L.O.)
| |
Collapse
|
14
|
Vooren EV, den Broeck FV, Mahieu Q, Geens E, Heetvelde MV, De Bruyne M, de Sompele SV, Uppal S, Poliakov E, Dhaenens CM, Gregory-Evans CY, Hoefsloot L, Gonzalez AI, Kohl S, Zuleger T, Demaret T, Dominant RPE65-p.(E519K) Consortium, Tuupanen S, Ruys J, Os LV, Platteau E, Jacob J, Vermeer S, Postelmans L, Dahan K, Maystadt I, Rasquin F, Thiadens AA, Stephenson KA, Sheri N, Smirnov V, MacDonald IM, Gregory-Evans K, Redmond TM, De Zaeytijd J, Leroy BP, Bauwens M, De Baere E. RPE65 variant p.(E519K) causes a novel dominant adult-onset maculopathy in 83 affected individuals. RESEARCH SQUARE 2025:rs.3.rs-5849564. [PMID: 40386434 PMCID: PMC12083654 DOI: 10.21203/rs.3.rs-5849564/v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Recessive RPE65-related retinopathy is an inherited retinal disease (IRD) that is a well-established target for gene therapy. Dominant RPE65-related retinopathy, however, due to Irish founder variant p.(D477G), is extremely rare. Here, we report the discovery, replication and characterization of a novel dominant retinopathy caused by RPE65 variant p.(E519K), identified in 83 individuals of European ancestry across IRD registries (Belgian discovery cohort, n=2,873; replication cohort, n=18,796). Long-read sequencing-based haplotyping revealed a shared region of 464 kb, supporting a founder effect. Genotype-phenotype data support dominant inheritance and phenotypic variability respectively, characterized by late-onset macular dystrophy with two main subtypes, a pathognomonic mottled subtype and a pattern dystrophy subtype. Functional studies showed that the p.(E519K) variant affects RPE65 enzymatic activity, correlating with lower protein expression. Protein modelling and cellular thermal shift assays further supported a destabilizing effect on protein structure. Overall, our work provides strong genetic, clinical, molecular and functional evidence for a novel dominant RPE65 retinopathy in multiple families in Europe and North America due to a Belgian founder variant. This discovery reduces the diagnostic gap in dominant IRD, particularly in individuals of European ancestry. Finally, it lays the foundation for developing therapeutic strategies targeting dominant RPE65 retinopathy.
Collapse
Affiliation(s)
- Eline Van Vooren
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Filip Van den Broeck
- Department of Head and Skin, Ghent University, Ghent, 9000, Belgium
- Department of Ophthalmology, Ghent University Hospital, Ghent, 9000, Belgium
| | - Quinten Mahieu
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Eline Geens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Mattias Van Heetvelde
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Marieke De Bruyne
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | | | - Sheetal Uppal
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, NIH, Bethesda, 20892, United States
| | - Eugenia Poliakov
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, NIH, Bethesda, 20892, United States
| | - Claire-Marie Dhaenens
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience & Cognition, F-59000 Lille, 59000, France
| | - Cheryl Y. Gregory-Evans
- University of British Columbia, Department of Ophthalmology & Visual Sciences, Vancouver, BC, V6T 1Z4, Canada
| | - Lies Hoefsloot
- Department of Clinical Genetics, Erasmus Medical Centre, Rotterdam, 3015, The Netherlands
| | | | - Susanne Kohl
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Hospital Tübingen, 72074, Germany
| | - Theresia Zuleger
- Institute of Medical Genetics and Applied Genomics, University Hospital Tübingen, 72074, Germany
| | - Tanguy Demaret
- Centre de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, 6041, Belgium
| | | | | | - Joke Ruys
- Department of Ophthalmology, Ghent University Hospital, Ghent, 9000, Belgium
- Department of Ophthalmology, Vitaz, Sint-Niklaas, 9100, Belgium
| | - Luc Van Os
- Department of Ophthalmology, Antwerp University Hospital, Antwerp, 2650, Belgium
| | - Elise Platteau
- Department Ophthalmology, Maria Middelares Hospital, Ghent, 9000, Belgium
| | - Julie Jacob
- Department of Ophthalmology, UZ Leuven, Leuven, 3000, Belgium
| | - Sascha Vermeer
- Center for Human Genetics, UZ Leuven, Leuven, 3000, Belgium
| | | | - Karin Dahan
- Centre de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, 6041, Belgium
| | - Isabelle Maystadt
- Centre de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, 6041, Belgium
| | - Florence Rasquin
- Department of ophthalmology, Erasme Hospital, Université Libre de Bruxelles, Brussels, 1070, Belgium
| | | | - Kirk A.J. Stephenson
- University of British Columbia, Department of Ophthalmology & Visual Sciences, Vancouver, BC, V6T 1Z4, Canada
| | - Narin Sheri
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Vasily Smirnov
- Univ. Lille, Inserm, CHU Lille, U1172-LilNCog-Lille Neuroscience & Cognition, F-59000 Lille, 59000, France
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, Paris, 75012, France
| | - Ian M. MacDonald
- Department of Ophthalmology and Visual Sciences, University of Alberta, Edmonton, T6G 2R3, Canada
| | - Kevin Gregory-Evans
- University of British Columbia, Department of Ophthalmology & Visual Sciences, Vancouver, BC, V6T 1Z4, Canada
| | - T. Michael Redmond
- Laboratory of Retinal Cell and Molecular Biology, National Eye Institute, NIH, Bethesda, 20892, United States
| | - Julie De Zaeytijd
- Department of Ophthalmology, Ghent University Hospital, Ghent, 9000, Belgium
| | - Bart P. Leroy
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
- Department of Head and Skin, Ghent University, Ghent, 9000, Belgium
- Department of Ophthalmology, Ghent University Hospital, Ghent, 9000, Belgium
- Division of Ophthalmology and Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA, 19104, United States
| | - Miriam Bauwens
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| | - Elfride De Baere
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- Center for Medical Genetics, Ghent University Hospital, Ghent, 9000, Belgium
| |
Collapse
|
15
|
Yamaguchi TN, Houlahan KE, Zhu H, Kurganovs N, Livingstone J, Fox NS, Yuan J, Sietsma Penington J, Jung CH, Schwarz T, Jaratlerdsiri W, van Riet J, Georgeson P, Mangiola S, Taraszka K, Lesurf R, Jiang J, Chow K, Heisler LE, Shiah YJ, Ramanand SG, Clarkson MJ, Nguyen A, Espiritu SMG, Stuchbery R, Jovelin R, Huang V, Bell C, O’Connor E, McCoy PJ, Lalansingh CM, Cmero M, Salcedo A, Chan EK, Liu LY, Stricker PD, Bhandari V, Bornman RM, Sendorek DH, Lonie A, Prokopec SD, Fraser M, Peters JS, Foucal A, Mutambirwa SB, Mcintosh L, Orain M, Wakefield M, Picard V, Park DJ, Hovington H, Kerger M, Bergeron A, Sabelnykova V, Seo JH, Pomerantz MM, Zaitlen N, Waszak SM, Gusev A, Lacombe L, Fradet Y, Ryan A, Kishan AU, Lolkema MP, Weischenfeldt J, Têtu B, Costello AJ, Hayes VM, Hung RJ, He HH, McPherson JD, Pasaniuc B, van der Kwast T, Papenfuss AT, Freedman ML, Pope BJ, Bristow RG, Mani RS, Corcoran NM, Reimand J, Hovens CM, Boutros PC. The Germline and Somatic Origins of Prostate Cancer Heterogeneity. Cancer Discov 2025; 15:988-1017. [PMID: 39945744 PMCID: PMC12046336 DOI: 10.1158/2159-8290.cd-23-0882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/06/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
SIGNIFICANCE This study uncovered 223 recurrently mutated driver regions using the largest cohort of prostate tumors to date. It reveals associations between germline SNPs, somatic drivers, and tumor aggression, offering significant insights into how prostate tumor evolution is shaped by germline factors and the timing of somatic mutations.
Collapse
Affiliation(s)
- Takafumi N. Yamaguchi
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
| | - Kathleen E. Houlahan
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute, Toronto, Canada
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Helen Zhu
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Natalie Kurganovs
- Ontario Institute for Cancer Research, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Julie Livingstone
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
| | - Natalie S. Fox
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Jiapei Yuan
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | | | - Chol-Hee Jung
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
| | - Tommer Schwarz
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, California
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California
| | - Weerachai Jaratlerdsiri
- Laboratory for Human Comparative and Prostate Cancer Genomics, Genomics and Epigenetics Division, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Job van Riet
- Department of Medical Oncology, Erasmus University, Rotterdam, the Netherlands
| | - Peter Georgeson
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
| | - Stefano Mangiola
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Bioinformatics Division, Walter and Eliza Hall Institute, Parkville, Australia
| | - Kodi Taraszka
- Department of Computer Science, University of California, Los Angeles, Los Angeles, California
| | - Robert Lesurf
- Ontario Institute for Cancer Research, Toronto, Canada
| | - Jue Jiang
- Laboratory for Human Comparative and Prostate Cancer Genomics, Genomics and Epigenetics Theme, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Ken Chow
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Division of Urology, Royal Melbourne Hospital, Parkville, Australia
| | | | - Yu-Jia Shiah
- Ontario Institute for Cancer Research, Toronto, Canada
| | | | - Michael J. Clarkson
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Anne Nguyen
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | | | - Ryan Stuchbery
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | | | - Vincent Huang
- Ontario Institute for Cancer Research, Toronto, Canada
| | - Connor Bell
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Edward O’Connor
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Patrick J. McCoy
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | | | - Marek Cmero
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Bioinformatics Division, Walter and Eliza Hall Institute, Parkville, Australia
| | - Adriana Salcedo
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Eva K.F. Chan
- St Vincent’s Clinical School, University of New South Wales, Randwick, Australia
- Department of Urology, St. Vincent’s Hospital Sydney, Darlinghurst, Australia
| | - Lydia Y. Liu
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute, Toronto, Canada
| | - Phillip D. Stricker
- Department of Urology, St. Vincent’s Hospital Sydney, Darlinghurst, Australia
| | - Vinayak Bhandari
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Riana M.S. Bornman
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
| | | | - Andrew Lonie
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
| | | | - Michael Fraser
- Ontario Institute for Cancer Research, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Justin S. Peters
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Adrien Foucal
- Ontario Institute for Cancer Research, Toronto, Canada
| | | | - Lachlan Mcintosh
- Bioinformatics Division, Walter and Eliza Hall Institute, Parkville, Australia
| | - Michèle Orain
- Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Matthew Wakefield
- Bioinformatics Division, Walter and Eliza Hall Institute, Parkville, Australia
| | - Valérie Picard
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Daniel J. Park
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
| | - Hélène Hovington
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Michael Kerger
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
| | - Alain Bergeron
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | | | - Ji-Heui Seo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mark M. Pomerantz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Noah Zaitlen
- Department of Neurology, University of California, Los Angeles, Los Angeles, California
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, California
| | - Sebastian M. Waszak
- Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo and Oslo University Hospital, Oslo, Norway
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Alexander Gusev
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Genetics, Brigham Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- The Eli and Edythe L. Broad Institute, Cambridge, Massachusetts
| | - Louis Lacombe
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Yves Fradet
- Division of Urology and Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Andrew Ryan
- TissuPath Specialist Pathology Services, Mount Waverley, Australia
| | - Amar U. Kishan
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Department of Radiation Oncology, University of California, Los Angeles, Los Angeles, California
| | - Martijn P. Lolkema
- Department of Computer Science, University of California, Los Angeles, Los Angeles, California
- Center for Personalized Cancer Treatment, Rotterdam, the Netherlands
| | - Joachim Weischenfeldt
- Biotech Research & Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
- Finsen Laboratory, Rigshospitalet, Copenhagen, Denmark
- Department of Urology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bernard Têtu
- Research Centre of CHU de Québec-Université Laval, Québec City, Canada
| | - Anthony J. Costello
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Division of Urology, Royal Melbourne Hospital, Parkville, Australia
| | - Vanessa M. Hayes
- St Vincent’s Clinical School, University of New South Wales, Randwick, Australia
- Department of Urology, St. Vincent’s Hospital Sydney, Darlinghurst, Australia
- School of Health Systems and Public Health, University of Pretoria, Pretoria, South Africa
- Central Clinical School, University of Sydney, Camperdown, Australia
- Department of Medical Sciences, University of Limpopo, Mankweng, South Africa
| | - Rayjean J. Hung
- Prosserman Centre for Population Health Research, Lunenfeld-Tanenbaum Research Institute, Toronto, Canada
- Epidemiology Division, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Housheng H. He
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - John D. McPherson
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Bogdan Pasaniuc
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, California
- Department of Computational Medicine, University of California, Los Angeles, Los Angeles, California
| | | | - Anthony T. Papenfuss
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
- Department of Mathematics and Statistics, University of Melbourne, Parkville, Australia
- Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | - Matthew L. Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Bernard J. Pope
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Melbourne Bioinformatics, The University of Melbourne, Melbourne, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, Australia
- Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
- Department of Medicine, Monash University, Clayton, Australia
| | - Robert G. Bristow
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
- Manchester Cancer Research Centre, Manchester, United Kingdom
| | - Ram S. Mani
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
- Department of Urology, UT Southwestern Medical Center, Dallas, Texas
| | - Niall M. Corcoran
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
- Division of Urology, Royal Melbourne Hospital, Parkville, Australia
- Department of Urology, Peninsula Health, Frankston, Australia
- The Victorian Comprehensive Cancer Centre, Parkville, Australia
| | - Jüri Reimand
- Ontario Institute for Cancer Research, Toronto, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Christopher M. Hovens
- Australian Prostate Cancer Research Centre Epworth, Richmond, Australia
- Department of Surgery, The University of Melbourne, Parkville, Australia
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Centre, University of California, Los Angeles, Los Angeles, California
- Institute for Precision Health, University of California, Los Angeles, Los Angeles, California
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute, Toronto, Canada
- Department of Urology, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
16
|
Ha YJJ, Nisal A, Tang I, Lee C, Jhamb I, Wallace C, Howarth R, Schroeder S, Vong KI, Meave N, Jiwani F, Barrows C, Lee S, Jiang N, Patel A, Bagga K, Banka N, Friedman L, Blanco FA, Yu S, Rhee S, Jeong HS, Plutzer I, Major MB, Benoit B, Poüs C, Heffner C, Kibar Z, Bot GM, Northrup H, Au KS, Strain M, Ashley-Koch AE, Finnell RH, Le JT, Meltzer HS, Araujo C, Machado HR, Stevenson RE, Yurrita A, Mumtaz S, Ahmed A, Khara MH, Mutchinick OM, Medina-Bereciartu JR, Hildebrandt F, Melikishvili G, Marwan AI, Capra V, Noureldeen MM, Salem AMS, Issa MY, Zaki MS, Xu L, Lee JE, Shin D, Alkelai A, Shuldiner AR, Kingsmore SF, Murray SA, Gee HY, Miller WT, Tolias KF, Wallingford JB, Kim S, Gleeson JG. The contribution of de novo coding mutations to meningomyelocele. Nature 2025; 641:419-426. [PMID: 40140573 DOI: 10.1038/s41586-025-08676-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 01/20/2025] [Indexed: 03/28/2025]
Abstract
Meningomyelocele (also known as spina bifida) is considered to be a genetically complex disease resulting from a failure of the neural tube to close. Individuals with meningomyelocele display neuromotor disability and frequent hydrocephalus, requiring ventricular shunting. A few genes have been proposed to contribute to disease susceptibility, but beyond that it remains unexplained1. We postulated that de novo mutations under purifying selection contribute to the risk of developing meningomyelocele2. Here we recruited a cohort of 851 meningomyelocele trios who required shunting at birth and 732 control trios, and found that de novo likely gene disruption or damaging missense mutations occurred in approximately 22.3% of subjects, with 28% of such variants estimated to contribute to disease risk. The 187 genes with damaging de novo mutations collectively define networks including actin cytoskeleton and microtubule-based processes, Netrin-1 signalling and chromatin-modifying enzymes. Gene validation demonstrated partial or complete loss of function, impaired signalling and defective closure of the neural tube in Xenopus embryos. Our results indicate that de novo mutations make key contributions to meningomyelocele risk, and highlight critical pathways required for neural tube closure in human embryogenesis.
Collapse
Affiliation(s)
- Yoo-Jin Jiny Ha
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ashna Nisal
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Isaac Tang
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Chanjae Lee
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Ishani Jhamb
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Cassidy Wallace
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Robyn Howarth
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Sarah Schroeder
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Keng Ioi Vong
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Naomi Meave
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Fiza Jiwani
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Chelsea Barrows
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Sangmoon Lee
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Nan Jiang
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Arzoo Patel
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Krisha Bagga
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Niyati Banka
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Liana Friedman
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
| | - Francisco A Blanco
- Department of Neuroscience, Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Seyoung Yu
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soeun Rhee
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Hui Su Jeong
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Isaac Plutzer
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, MO, USA
| | - Michael B Major
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, MO, USA
| | - Béatrice Benoit
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
| | - Christian Poüs
- INSERM UMR-S 1193, UFR de Pharmacie, University Paris-Saclay, Orsay, France
- Biochimie-Hormonologie, Assistance Publique - Hôpitaux de Paris Université Paris-Saclay, Clamart, France
| | | | - Zoha Kibar
- Department of Neurosciences, Research Center of CHU Sainte Justine, University of Montreal, Montreal, Quebec, Canada
| | - Gyang Markus Bot
- Neurosurgery Division, Department of Surgery, Jos University Teaching Hospital, Jos, Nigeria
| | - Hope Northrup
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Kit Sing Au
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center at Houston and Children's Memorial Hermann Hospital, Houston, TX, USA
| | - Madison Strain
- Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | | | - Richard H Finnell
- Center for Precision Environmental Health, Departments of Molecular and Human Genetics, Molecular and Cellular Biology and Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Joan T Le
- Rady Children's Hospital, San Diego, CA, USA
| | | | - Camila Araujo
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Helio R Machado
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Roger E Stevenson
- J. C. Self Research Institute of Human Genetics, Greenwood Genetic Center, Greenwood, SC, USA
| | - Anna Yurrita
- Catedrática de Ciencias Ómicas, Facultad de Medicina, Universidad Francisco Marroquín, Guatemala City, Guatemala
| | - Sara Mumtaz
- National University of Medical Sciences, Rawalpindi, Pakistan
| | | | | | - Osvaldo M Mutchinick
- Department of Genetics, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | | | | | - Gia Melikishvili
- Department of Pediatrics, MediClubGeorgia Medical Center, Tbilisi, Georgia
| | - Ahmed I Marwan
- Division of Pediatric General, Thoracic and Fetal Surgery, Department of Surgery, University of Missouri School of Medicine, Columbia, MO, USA
| | - Valeria Capra
- Genomics and Clinical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Mahmoud M Noureldeen
- Department of Pediatrics, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Aida M S Salem
- Department of Pediatrics, Faculty of Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Mahmoud Y Issa
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Cairo, Egypt
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Ji Eun Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Donghyuk Shin
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | | | | | | | | | - Heon Yung Gee
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - W Todd Miller
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
- VA Medical Center, Northport, NY, USA
| | - Kimberley F Tolias
- Department of Neuroscience, Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, USA
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Sangwoo Kim
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- POSTECH Biotechnology Center, Pohang University of Science and Technology, Pohang, Republic of Korea.
| | - Joseph G Gleeson
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA.
- Department of Neurosciences and Pediatrics, University of California, San Diego, San Diego, CA, USA.
| |
Collapse
|
17
|
Abstract
A single reference genome does not fully capture species diversity. By contrast, a pangenome incorporates multiple genomes to capture the entire set of nonredundant genes in a given species, along with its genome diversity. New sequencing technologies enable researchers to produce multiple high-quality genome sequences and catalog diverse genetic variations with better precision. Pangenomic studies have detected structural variants in plant genomes, dissected the genetic architecture of agronomic traits, and helped unravel molecular underpinnings and evolutionary origins of plant phenotypes. The pangenome concept has further evolved into a so-called super-pangenome that includes wild relatives within a genus or clade and shifted to graph-based reference systems. Nevertheless, building pangenomes and representing complex structural variants remain challenging in many crops. Standardized computing pipelines and common data structures are needed to compare and interpret pangenomes. The growing body of plant pangenomics data requires new algorithms, huge data storage capacity, and training to help researchers and breeders take advantage of newly discovered genes and genetic variants.
Collapse
Affiliation(s)
- Murukarthick Jayakodi
- Department of Soil and Crop Sciences, Texas A&M University, College Station, Texas, USA;
- Texas A&M AgriLife Research Center at Dallas, Texas A&M University System, Dallas, Texas, USA
| | - Hyeonah Shim
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Seeland, Germany
| | - Martin Mascher
- German Centre for Integrative Biodiversity Research (iDiv), Halle-Jena-Leipzig, Leipzig, Germany;
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Seeland, Germany
| |
Collapse
|
18
|
Wang Y, Li H, Li Q, Li Y, Wu H, Ge Y, Zhu X, Zheng Z, Sun Z. Characterization of Extrachromosomal Circular DNA in Primary and Cisplatin-Resistant High-Grade Serous Ovarian Cancer. Genes (Basel) 2025; 16:517. [PMID: 40428339 PMCID: PMC12111702 DOI: 10.3390/genes16050517] [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: 03/20/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Cisplatin resistance is a major cause of tumor recurrence and mortality in high-grade serous ovarian cancer (HGSOC). Extrachromosomal circular DNA (eccDNA) has emerged as a critical factor in tumor evolution and drug resistance. However, the specific contribution of eccDNA to cisplatin resistance in HGSOC remains unclear. METHODS We performed whole-genome sequencing, Circle-Seq, and RNA-Seq in four pairs of primary and cisplatin-resistant (cisR) HGSOC cell lines to characterize genome-wide eccDNA distribution and features. Functional enrichment analyses were subsequently conducted on differentially expressed eccDNA-related genes. RESULTS In the SKOV3 cisR cell line, we identified a large extrachromosomal circular DNA (ecDNA) carrying the HIF1A gene, which regulates DNA repair, drug efflux, and epithelial-mesenchymal transition, contributing to cisplatin resistance. Using Circle-Seq, we detected a total of 161,062 eccDNAs, most of which were less than 1000 bp and distributed across all chromosomes. Notably, the number of eccDNAs on chromosome 21 differed significantly between the primary and cisR cell lines. Additionally, eccDNAs were predominantly located in non-coding repetitive elements. Functional analysis of eccDNA-related differentially expressed genes revealed that, compared to primary cell lines, cisR cell lines were associated with mitotic spindle assembly, regulation of vascular permeability, and cell differentiation. eccDNA-related genes involved in these pathways include MISP, WIPF1, RHOD, KRT80, and PLVAP. CONCLUSIONS Our findings suggest that eccDNAs, particularly ecDNA amplifications like HIF1A, contribute significantly to cisplatin resistance mechanisms in HGSOC. These insights highlight eccDNA as a potential target for overcoming therapeutic resistance and improving treatment outcomes in ovarian cancer.
Collapse
Affiliation(s)
- Youya Wang
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (H.L.); (H.W.); (Y.G.); (X.Z.)
| | - He Li
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (H.L.); (H.W.); (Y.G.); (X.Z.)
| | - Qinglan Li
- Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (Q.L.); (Y.L.)
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Yi Li
- Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (Q.L.); (Y.L.)
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing 100049, China
| | - Hao Wu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (H.L.); (H.W.); (Y.G.); (X.Z.)
| | - Yan Ge
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (H.L.); (H.W.); (Y.G.); (X.Z.)
| | - Xingnuo Zhu
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (H.L.); (H.W.); (Y.G.); (X.Z.)
| | - Zhiguo Zheng
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Zhongsheng Sun
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou 325000, China; (Y.W.); (H.L.); (H.W.); (Y.G.); (X.Z.)
- Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; (Q.L.); (Y.L.)
- College of Life Sciences, University of the Chinese Academy of Sciences, Beijing 100049, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| |
Collapse
|
19
|
Butt RN, Amina B, Sultan MU, Tanveer ZB, Gondal MN, Hussain R, Khan S, Akbar R, Nasir Z, Khalid MF, Channan-Khan AA, Faisal A, Shoaib M, Chaudhary SU. CanSeer: a translational methodology for developing personalized cancer models and therapeutics. Sci Rep 2025; 15:15080. [PMID: 40301468 PMCID: PMC12041273 DOI: 10.1038/s41598-025-99219-x] [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: 07/13/2024] [Accepted: 04/16/2025] [Indexed: 05/01/2025] Open
Abstract
Computational modeling and analysis of biomolecular network models annotated with omics data are emerging as a versatile tool for designing personalized therapies. Current endeavors aimed at employing in silico models towards personalized cancer therapeutics remain limited in providing all-in-one approach that ascertains actionable targets, re-positions FDA (Food and Drug Administration) approved drugs, furnishes quantitative cues on therapy responses such as efficacy and cytotoxic effect, and identifies novel drug combinations. Here we propose "CanSeer"-a methodology for developing personalized therapeutics. CanSeer employs patient-specific genetic alterations and RNA-seq data to annotate in silico models followed by dynamical network analyses towards assessment of treatment responses. To exemplify, three use cases involving paired samples, unpaired samples, and cancer samples only, of lung squamous cell carcinoma (LUSC) patients are provided. CanSeer reveals the effectiveness of repositioned drugs along with the identification of several novel LUSC treatment combinations including Afuresertib + Palbociclib, Dinaciclib + Trametinib, Afatinib + Oxaliplatin, Ulixertinib + Olaparib, etc.
Collapse
Affiliation(s)
- Rida Nasir Butt
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Bibi Amina
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Muhammad Umer Sultan
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Zain Bin Tanveer
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Mahnoor Naseer Gondal
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Risham Hussain
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
- Data Science Institute, Lancaster University, Lancaster, LA1 4YW, UK
| | - Salaar Khan
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
- Center for Eukaryotic Gene Regulation, Department of Biochemistry and Molecular Biology, Penn State University, University Park, PA, 16802, USA
| | - Rida Akbar
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Zainab Nasir
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Muhammad Farhan Khalid
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | | | - Amir Faisal
- Cancer Therapeutics Lab, Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Muhammad Shoaib
- Epigenome and Genome Integrity Lab (EaGIL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan
| | - Safee Ullah Chaudhary
- Biomedical Informatics and Engineering Research Laboratory (BIRL), Syed Babar Ali School of Science and Engineering, Department of Life Sciences, Lahore University of Management Sciences, Lahore, 54792, Pakistan.
| |
Collapse
|
20
|
Li Y, Xu X, She Y, Su Z, Liu X, Chen Y, Ye C, Zhang Y, Yu H, Chen C, Chen S, Zhou L. Genetic Etiology of Epilepsy: A Retrospective Study From a Single-Center Cohort. Clin Genet 2025. [PMID: 40296287 DOI: 10.1111/cge.14757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025]
Abstract
Next generation sequencing (NGS) technology has made significant progress in the genetic diagnosis and treatment of epilepsy. However, genetic studies on epilepsy with different etiologies remain relatively limited. In this study, whole-genome or whole-exome sequencing was performed on 158 unrelated patients with epilepsy of various etiologies, and the identified variants were analyzed for their association with 1356 seizure-related genes in the database. Additionally, the pathogenicity or likely pathogenicity of those variants associated with known epilepsy genes was evaluated. The results showed that pathogenic or likely pathogenic variants were detected in 31.65% (50/158) of the patients in our cohort study. Further analysis revealed significant differences in the diagnostic rates among different epilepsy categories: 29.60% (37/125) for idiopathic epilepsy and 39.39% (13/33) for symptomatic epilepsy. Moreover, the genes PRRT2, KMT2C, PRKRA, NOTCH3, NAGLU, and SCN1A were identified as potentially important for epilepsy, suggesting they could become key targets for clinical diagnosis and treatment. In conclusion, NGS technology demonstrates high diagnostic efficiency for epilepsy of different etiologies and highlights significant differences among various types. This provides novel genetic insights for the diagnosis and treatment of epilepsy.
Collapse
Affiliation(s)
- Yinchao Li
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Pediatrics, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xiaowei Xu
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yingfang She
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhengwei Su
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xianyue Liu
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Ying Chen
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chenghui Ye
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yuanchao Zhang
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Hang Yu
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chun Chen
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Shuda Chen
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Liemin Zhou
- Department of Neurology, The Seven Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
21
|
Patel Y, Zhu C, Yamaguchi TN, Wang NK, Wiltsie N, Zeltser N, Gonzalez AE, Winata HK, Pan Y, Mootor MFE, Sanders T, Fitz-Gibbon ST, Kandoth C, Livingstone J, Liu LY, Carlin B, Holmes A, Oh J, Sahrmann J, Tao S, Eng S, Hugh-White R, Pashminehazar K, Park A, Beshlikyan A, Jordan M, Wu S, Tian M, Arbet J, Neilsen B, Haas R, Bugh YZ, Kim G, Salmingo J, Zhang W, Anand A, Hwang E, Neiman-Golden A, Steinberg P, Zhao W, Anand P, Agrawal R, Tsai BL, Boutros PC. Metapipeline-DNA: A Comprehensive Germline & Somatic Genomics Nextflow Pipeline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.04.611267. [PMID: 39282325 PMCID: PMC11398472 DOI: 10.1101/2024.09.04.611267] [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: 09/21/2024]
Abstract
Summary The price, quality and throughout of DNA sequencing continue to improve. Algorithmic innovations have allowed inference of a growing range of features from DNA sequencing data, quantifying nuclear, mitochondrial and evolutionary aspects of both germline and somatic genomes. To automate analyses of the full range of genomic characteristics, we created an extensible Nextflow meta-pipeline called metapipeline-DNA. Metapipeline-DNA analyzes targeted and whole-genome sequencing data from raw reads through pre-processing, feature detection by multiple algorithms, quality-control and data- visualization. Each step can be run independently and is supported robust software engineering including automated failure-recovery, robust testing and consistent verifications of inputs, outputs and parameters. Metapipeline-DNA is cloud-compatible and highly configurable, with options to subset and optimize each analysis. Metapipeline-DNA facilitates high-scale, comprehensive analysis of DNA sequencing data. Availability Metapipeline-DNA is an open-source Nextflow pipeline under the GPLv2 license and is available at https://github.com/uclahs-cds/metapipeline-DNA .
Collapse
|
22
|
Ortiz-Martínez D. DeBasher: a flow-based programming bash extension for the implementation of complex and interactive workflows with stateful processes. BMC Bioinformatics 2025; 26:106. [PMID: 40240939 PMCID: PMC12004750 DOI: 10.1186/s12859-025-06108-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/07/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Bioinformatics data analysis faces significant challenges. As data analysis often takes the form of pipelines or workflows, workflow managers (WfMs) have become essential. Data flow programming constitutes the preferred approach in WfMs, enabling parallel processes activated reactively based on input availability. While this paradigm typically follows a linear, acyclic progression, cyclic workflows are sometimes necessary in bioinformatics analyses. These cyclic workflows also present an opportunity to explore workflow interactivity, a feature not widely implemented in existing WfMs. RESULTS We propose DeBasher, a tool that adopts the flow-based programming (FBP) paradigm, in which the workflow components are in control of their life cycle and can store state information, allowing the execution of complex workflows that include cycles. DeBasher also incorporates a powerful model of interactivity, where the user can alter the behavior of a running workflow. Additionally, DeBasher allows the user to define triggers so as to initiate the execution of a complete workflow or a part of it. The ability to execute processes with state and in control of their life cycle also has applications in dynamic scheduling tasks. Furthermore, DeBasher presents a series of extra features, including the combination of multiple workflows at runtime through a feature we have called runtime piping, switching to static scheduling to increase scalability, or implementing processes in multiple languages. DeBasher has been successfully used to process 131.7 TB of genomic data by means of a variant calling pipeline. CONCLUSIONS DeBasher is an FBP Bash extension that can be useful in a wide range of situations and in particular when implementing complex workflows, workflows with interactivity or triggers, or when a high scalability is required.
Collapse
Affiliation(s)
- Daniel Ortiz-Martínez
- Department of Mathematics and Computer Science, Universitat de Barcelona, Gran Via de les Corts Catalanes, 585, 08007, Barcelona, Spain.
| |
Collapse
|
23
|
Mahmoud M, Agustinho DP, Sedlazeck FJ. A Hitchhiker's Guide to long-read genomic analysis. Genome Res 2025; 35:545-558. [PMID: 40228901 PMCID: PMC12047252 DOI: 10.1101/gr.279975.124] [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] [Indexed: 04/16/2025]
Abstract
Over the past decade, long-read sequencing has evolved into a pivotal technology for uncovering the hidden and complex regions of the genome. Significant cost efficiency, scalability, and accuracy advancements have driven this evolution. Concurrently, novel analytical methods have emerged to harness the full potential of long reads. These advancements have enabled milestones such as the first fully completed human genome, enhanced identification and understanding of complex genomic variants, and deeper insights into the interplay between epigenetics and genomic variation. This mini-review provides a comprehensive overview of the latest developments in long-read DNA sequencing analysis, encompassing reference-based and de novo assembly approaches. We explore the entire workflow, from initial data processing to variant calling and annotation, focusing on how these methods improve our ability to interpret a wide array of genomic variants. Additionally, we discuss the current challenges, limitations, and future directions in the field, offering a detailed examination of the state-of-the-art bioinformatics methods for long-read sequencing.
Collapse
Affiliation(s)
- Medhat Mahmoud
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Daniel P Agustinho
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA;
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Computer Science, Rice University, Houston, Texas 77005, USA
| |
Collapse
|
24
|
Oggenfuss U, Todd RT, Soisangwan N, Kemp B, Guyer A, Beach A, Selmecki A. Candida albicans isolates contain frequent heterozygous structural variants and transposable elements within genes and centromeres. Genome Res 2025; 35:824-838. [PMID: 39438112 PMCID: PMC12047244 DOI: 10.1101/gr.279301.124] [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: 03/09/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024]
Abstract
The human fungal pathogen Candida albicans poses a significant burden on global health, causing high rates of mortality and antifungal drug resistance. C. albicans is a heterozygous diploid organism that reproduces asexually. Structural variants (SVs) are an important source of genomic rearrangement, particularly in species that lack sexual recombination. To comprehensively investigate SVs across clinical isolates of C. albicans, we conducted long-read sequencing and genome-wide SV analysis in three distantly related clinical isolates. Our work includes a new, comprehensive analysis of transposable element (TE) composition, location, and diversity. SVs and TEs are frequently close to coding sequences and many SVs are heterozygous, suggesting that SVs might impact gene and allele-specific expression. Most SVs are uniquely present in only one clinical isolate, indicating that SVs represent a significant source of intraspecies genetic variation. We identify multiple, distinct SVs at the centromeres of Chromosome 4 and Chromosome 5, including inversions and transposon polymorphisms. These two chromosomes are often aneuploid in drug-resistant clinical isolates and can form isochromosome structures with breakpoints near the centromere. Further screening of 100 clinical isolates confirms the widespread presence of centromeric SVs in C. albicans, often appearing in a heterozygous state, indicating that SVs are contributing to centromere evolution in C. albicans Together, these findings highlight that SVs and TEs are common across diverse clinical isolates of C. albicans and that the centromeres of this organism are important sites of genome rearrangement.
Collapse
Affiliation(s)
- Ursula Oggenfuss
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Robert T Todd
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55455, USA
- Department of Biology, Bard College, Annandale-on-Hudson, New York 12504, USA
| | - Natthapon Soisangwan
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Bailey Kemp
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Alison Guyer
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Annette Beach
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Anna Selmecki
- Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota 55455, USA;
| |
Collapse
|
25
|
De La Vega FM, Irvine SA, Anur P, Potts K, Kraft L, Torres R, Kang P, Truong S, Lee Y, Han S, Onuchic V, Han J. Benchmarking of germline copy number variant callers from whole genome sequencing data for clinical applications. BIOINFORMATICS ADVANCES 2025; 5:vbaf071. [PMID: 40248358 PMCID: PMC12005901 DOI: 10.1093/bioadv/vbaf071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 03/11/2025] [Accepted: 04/08/2025] [Indexed: 04/19/2025]
Abstract
Motivation Whole-genome sequencing (WGS) is increasingly preferred for clinical applications due to its comprehensive coverage, effectiveness in detecting copy number variants (CNVs), and declining costs. However, systematic evaluations of WGS CNV callers tailored to germline clinical testing-where high sensitivity and confirmation of reported CNVs are essential-remain necessary. Clinical reporting typically emphasizes CNVs affecting coding regions over precise breakpoint detection. This study benchmarks several short-read WGS CNV detection tools using reference cell lines to inform their clinical use. Results While tools vary in sensitivity (7%-83%) and precision (1%-76%), few meet the sensitivity needed for clinical testing. Callers generally perform better for deletions (up to 88% sensitivity) than duplications (up to 47% sensitivity), with poor detection of duplications under 5 kb. Notably, for CNVs in genes commonly included in clinical panels, significantly improved sensitivity and precision were observed when benchmarking against 25 cell lines with known CNVs. DRAGEN v4.2 high-sensitivity CNV calls, post-processed with custom filters, achieved 100% sensitivity and 77% precision on the optimized gene panel after excluding recurring artifacts. This level of performance may support clinical use with orthogonal confirmation of reportable CNVs, pending validation on laboratory-specific samples. Availability and implementation The data underlying this article are available in the European Nucleo-tide Archive under project accession PRJEB87628.
Collapse
Affiliation(s)
- Francisco M De La Vega
- Tempus AI, Inc., Chicago, IL 60654, United States
- Department of Biomedical Data Sciences, Stanford University School of Medicine, Palo Alto, CA 94304, United States
| | - Sean A Irvine
- Real Time Genomics, Ltd., Hamilton 3204, New Zealand
| | - Pavana Anur
- Tempus AI, Inc., Chicago, IL 60654, United States
| | - Kelly Potts
- Tempus AI, Inc., Chicago, IL 60654, United States
| | - Lewis Kraft
- Tempus AI, Inc., Chicago, IL 60654, United States
| | - Raul Torres
- Tempus AI, Inc., Chicago, IL 60654, United States
| | - Peter Kang
- Tempus AI, Inc., Chicago, IL 60654, United States
| | - Sean Truong
- llumina, Inc., San Diego, CA 92122, United States
| | - Yeonghun Lee
- llumina, Inc., San Diego, CA 92122, United States
| | - Shunhua Han
- llumina, Inc., San Diego, CA 92122, United States
| | | | - James Han
- llumina, Inc., San Diego, CA 92122, United States
| |
Collapse
|
26
|
Xu IRL, Danzi MC, Raposo J, Züchner S. The continued promise of genomic technologies and software in neurogenetics. J Neuromuscul Dis 2025:22143602251325345. [PMID: 40208247 DOI: 10.1177/22143602251325345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
The continued evolution of genomic technologies over the past few decades has revolutionized the field of neurogenetics, offering profound insights into the genetic underpinnings of neurological disorders. Identification of causal genes for numerous monogenic neurological conditions has informed key aspects of disease mechanisms and facilitated research into critical proteins and molecular pathways, laying the groundwork for therapeutic interventions. However, the question remains: has this transformative trend reached its zenith? In this review, we suggest that despite significant strides in genome sequencing and advanced computational analyses, there is still ample room for methodological refinement. We anticipate further major genetic breakthroughs corresponding with the increased use of long-read genomes, variant calling software, AI tools, and data aggregation databases. Genetic progress has historically been driven by technological advancements from the commercial sector, which are developed in response to academic research needs, creating a continuous cycle of innovation and discovery. This review explores the potential of genomic technologies to address the challenges of neurogenetic disorders. By outlining both established and modern resources, we aim to emphasize the importance of genetic technologies as we enter an era poised for discoveries.
Collapse
Affiliation(s)
- Isaac R L Xu
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Matt C Danzi
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jacquelyn Raposo
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stephan Züchner
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
27
|
Zhang X, Bai Q, Wang Y, Jiang Z, Han J, Xue C, Huang K, Luan L, Huang X, Huang X, Shi G, Hou Y, Ji Y. FGFR2 fusion/rearrangement analysis in intrahepatic cholangiocarcinoma using DNA/RNA-based NGS and FISH. Virchows Arch 2025:10.1007/s00428-025-04067-9. [PMID: 40198372 DOI: 10.1007/s00428-025-04067-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/04/2025] [Accepted: 02/22/2025] [Indexed: 04/10/2025]
Abstract
Patients with intrahepatic cholangiocarcinoma (iCCA) harboring FGFR2 fusion/rearrangement benefit from targeted therapies, highlighting the need for reliable testing strategies to identify FGFR2 alterations. We assessed 226 iCCA cases using RNA-based NGS, DNA-based NGS, and break-apart FISH to evaluate the effectiveness of these methods in detecting FGFR2 fusion/rearrangement. The detection rates for FGFR2 fusion/rearrangement were 9.7% (22/226) for RNA-based NGS, 7.1% (16/226) for DNA-based NGS, and 10.2% (23/226) for FISH. Among the 26 FGFR2 fusion/rearrangement-positive cases identified by any method, only 15 (57.7%) were positive by all three techniques, yielding a concordance rate of 95.1% (215/226). RNA-based NGS confirmed oncogenic FGFR2 fusion in 81% (21/26) of positive cases and identified five novel oncogenic fusions. Thirty-five percent (6/17) of the partner genes were located on chromosome 10, with BICC1 being the most common fusion partner, while the rest were distributed across the other 9 chromosomes. FISH demonstrated a sensitivity of 95.2% and specificity of 98.5%, compared to oncogenic FGFR2 fusions confirmed by RNA-based NGS, while DNA-based NGS exhibited a sensitivity of 71.4% and specificity of 99.5%, identifying FGFR2 mutations in 4 cases. FGFR2-FISH positive cases displayed no significant heterogeneity in positive cell distribution. Oncogenic FGFR2 fusion/rearrangement was associated with small duct type iCCA, especially in cases with positive serum HBsAg and absent cholangiolocarcinoma components and peripheral liver steatosis. This study provides a comprehensive comparison of three assays for detecting FGFR2 fusion/rearrangement, along with clinicopathologic characterization of oncogenic FGFR2 fusion in iCCA.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pathology, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Qianming Bai
- Department of Pathology, Fudan University Shanghai Cancer Centre, 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Yulin Wang
- Department of Pathology, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhengzeng Jiang
- Department of Pathology, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Jing Han
- Department of Pathology, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Cheng Xue
- Shanghai Epione Medlab Co., Ltd, 1158 Zhongxin Road, Shanghai, 201615, China
| | - Kai Huang
- Shanghai Epione Medlab Co., Ltd, 1158 Zhongxin Road, Shanghai, 201615, China
| | - Lijuan Luan
- Department of Pathology, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Xiaoyong Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Xiaowu Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Guoming Shi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Yingyong Hou
- Department of Pathology, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China
| | - Yuan Ji
- Department of Pathology, Fudan University Zhongshan Hospital, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
28
|
Ismail FN, Amarasoma S. Mars: simplifying bioinformatics workflows through a containerized approach to tool integration and management. BIOINFORMATICS ADVANCES 2025; 5:vbaf074. [PMID: 40406670 PMCID: PMC12095131 DOI: 10.1093/bioadv/vbaf074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/14/2025] [Accepted: 04/02/2025] [Indexed: 05/26/2025]
Abstract
Summary Bioinformatics is a rapidly evolving field with numerous specialized tools developed for essential genomic analysis tasks, such as read simulation, mapping, and variant calling. However, managing these tools presents significant challenges due to varied dependencies, execution steps, and output formats, complicating the installation and configuration processes. To address these issues, we introduce "Mars" a bioinformatics solution encapsulated within a singularity container that preloads a comprehensive suite of widely used genomic tools. Mars not only simplifies the installation of these tools but also automates critical workflow functions, including sequence sample preparation, read simulation, read mapping, variant calling, and result comparison. By streamlining the execution of these workflows, Mars enables users to easily manage input-output formats and compare results across different tools, thereby enhancing reproducibility and efficiency. Furthermore, by providing a cohesive environment that integrates tool management with a flexible workflow interface, Mars empowers researchers to focus on their analyses rather than the complexities of tool configuration. This integrated solution facilitates the testing of various combinations of tools and algorithms, enabling users to evaluate performance based on different metrics and identify the optimal tools for their specific genomic analysis needs. Through Mars, we aim to enhance the accessibility and usability of bioinformatics tools, ultimately advancing research in genomic analysis. Availability and implementation Mars is freely available at https://github.com/GenomicAI/mars. It is implemented within a Singularity container environment and supports modular extension for additional genomic tools and custom workflows.
Collapse
Affiliation(s)
- Fathima Nuzla Ismail
- Department of Mathematics, State University of New York at Buffalo, Buffalo, NY 14260, United States
| | - Shanika Amarasoma
- Independent Researcher, AI & Advanced Analytics, Colombo 01100, Sri Lanka
| |
Collapse
|
29
|
Farooq AR, Zhang AX, Chan-Seng-Yue M, Topham JT, O'Kane GM, Jang GH, Fischer S, Dodd A, Holter S, Wilson J, Grant RC, Aung KL, Zogopoulos G, Elimova E, Prince R, Jang R, Moore M, Biagi J, Tang P, Goodwin R, Bathe OF, Marra M, Laskin J, Renouf DJ, Schaeffer DF, Karasinska JM, Notta F, Gallinger S, Knox JJ, Tsang ES. The tandem duplicator phenotype may be a novel targetable subgroup in pancreatic cancer. NPJ Precis Oncol 2025; 9:100. [PMID: 40185871 PMCID: PMC11971333 DOI: 10.1038/s41698-025-00888-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Tandem duplicator phenotype (TDP) consists of distinct genomic rearrangements where tandem duplications are randomly distributed. In this study, we characterized the prevalence and outcomes of TDP in a large series of prospectively sequenced tumors from patients with pancreatic ductal adenocarcinomas (PDAC). Whole-genome sequencing (WGS) was performed in 530 PDAC cases from the PanCuRx Initiative, COMPASS and PanGen/POG trials in Canada. Of 530 cases, 52 were identified as TDP (9.8%; 13 resected, 39 advanced). Etiological subgroups of TDP included BRCA1 (n = 9), CCNE1 (n = 4), and unknown (n = 39). Presence of TDP was not prognostic in resected specimens (p = 0.77) compared with non-HRD and non-TDP cases, described as typicals. In advanced cases, when stratified for only classical subtype cases, platinum therapy was correlated with longer response in non-BRCA1 TDP vs. typicals (p = 0.0036). There was no difference in overall survival between TDP and typicals (p = 0.5).TDP represents a potential novel targetable subgroup for chemotherapy selection in PDAC.
Collapse
Affiliation(s)
| | - Amy X Zhang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | - James T Topham
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | | | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | - Anna Dodd
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Spring Holter
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Julie Wilson
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | | | | | - Elena Elimova
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - Raymond Jang
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Malcolm Moore
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - James Biagi
- Cancer Centre of Southeastern Ontario/Queen's University, Kingston, ON, Canada
| | - Patricia Tang
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Rachel Goodwin
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital, The University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Oliver F Bathe
- Department of Oncology, Tom Baker Cancer Center, University of Calgary, 1331 29th St NW, Calgary, AB, T2N 4N2, Canada
| | - Marco Marra
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Janessa Laskin
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Daniel J Renouf
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - David F Schaeffer
- Department of Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| | | | - Faiyaz Notta
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | | | - Erica S Tsang
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
30
|
Keskus AG, Bryant A, Ahmad T, Yoo B, Aganezov S, Goretsky A, Donmez A, Lansdon LA, Rodriguez I, Park J, Liu Y, Cui X, Gardner J, McNulty B, Sacco S, Shetty J, Zhao Y, Tran B, Narzisi G, Helland A, Cook DE, Chang PC, Kolesnikov A, Carroll A, Molloy EK, Bi C, Walter A, Gibson M, Pushel I, Guest E, Pastinen T, Shafin K, Miga KH, Malikic S, Day CP, Robine N, Sahinalp C, Dean M, Farooqi MS, Paten B, Kolmogorov M. Severus detects somatic structural variation and complex rearrangements in cancer genomes using long-read sequencing. Nat Biotechnol 2025:10.1038/s41587-025-02618-8. [PMID: 40185952 DOI: 10.1038/s41587-025-02618-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/26/2025] [Indexed: 04/07/2025]
Abstract
For the detection of somatic structural variation (SV) in cancer genomes, long-read sequencing is advantageous over short-read sequencing with respect to mappability and variant phasing. However, most current long-read SV detection methods are not developed for the analysis of tumor genomes characterized by complex rearrangements and heterogeneity. Here, we present Severus, a breakpoint graph-based algorithm for somatic SV calling from long-read cancer sequencing. Severus works with matching normal samples, supports unbalanced cancer karyotypes, can characterize complex multibreak SV patterns and produces haplotype-specific calls. On a comprehensive multitechnology cell line panel, Severus consistently outperforms other long-read and short-read methods in terms of SV detection F1 score (harmonic mean of the precision and recall). We also illustrate that compared to long-read methods, short-read sequencing systematically misses certain classes of somatic SVs, such as insertions or clustered rearrangements. We apply Severus to several clinical cases of pediatric leukemia/lymphoma, revealing clinically relevant cryptic rearrangements missed by standard genomic panels.
Collapse
Affiliation(s)
- Ayse G Keskus
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Asher Bryant
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Tanveer Ahmad
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Byunggil Yoo
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | | | - Anton Goretsky
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Ataberk Donmez
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Lisa A Lansdon
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Isabel Rodriguez
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Jimin Park
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Yuelin Liu
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Xiwen Cui
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Joshua Gardner
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Brandy McNulty
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Samuel Sacco
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Jyoti Shetty
- Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Yongmei Zhao
- Sequencing Facility Bioinformatics Group, Biomedical Informatics and Data Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bao Tran
- Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | | | | | | | | | - Erin K Molloy
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Chengpeng Bi
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Adam Walter
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Margaret Gibson
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Irina Pushel
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Erin Guest
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Tomi Pastinen
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Kishwar Shafin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Karen H Miga
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Salem Malikic
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Chi-Ping Day
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Cenk Sahinalp
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Michael Dean
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Midhat S Farooqi
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Benedict Paten
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Mikhail Kolmogorov
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
31
|
Pan T, Miao J, Sun K, Nie H, Luscombe NM, Li W, Zhang S, Yang L, Wang H, Zhou Y, Tu G, Shu Y, Zhang B, Wu X. Genomic insights and the conservation potential of captive breeding: The case of Chinese alligator. SCIENCE ADVANCES 2025; 11:eadm7980. [PMID: 40173227 PMCID: PMC11963981 DOI: 10.1126/sciadv.adm7980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 02/27/2025] [Indexed: 04/04/2025]
Abstract
Despite 40 years of conservation of the critically endangered Chinese alligator (Alligator sinensis), the genomic underpinnings of its status remained uncharted. Genome sequencing data of 244 individuals uncovered relatively low overall genomic diversity/heterozygosity and long runs of homozygosity, with captive populations exhibiting higher heterozygosity and smaller inbreeding coefficients compared to wild individuals. The decreased level of inbreeding in the captive population demonstrates the contribution of the large captive breeding population. The estimated recent effective population size was around a few dozen. To combat challenges of inbreeding depression and reduced adaptability, we used genome-wide SNP-based kinship analysis on captive populations to enable a genome-informed breeding program that minimizes inbreeding. Long-term field monitoring revealed that the Chinese government greatly advanced the conservation of A. sinensis through conservation measures and reintroduction programs. Our research enriches the understanding of the Chinese alligator's genetic landscape, offering invaluable genomic resources for breeding and conservation strategies.
Collapse
Affiliation(s)
- Tao Pan
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, Wuhu, Anhui 241000, China
| | - Jiashun Miao
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, Wuhu, Anhui 241000, China
- Genomics and Regulatory Systems Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan
- Xianghu Laboratory, Hangzhou, Zhejiang 311231, China
| | - Ke Sun
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, Wuhu, Anhui 241000, China
| | - Haitao Nie
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, Wuhu, Anhui 241000, China
| | - Nicholas M. Luscombe
- Genomics and Regulatory Systems Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa 904-0495, Japan
| | - Wengang Li
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, Wuhu, Anhui 241000, China
| | - Song Zhang
- Anhui Research Center of Chinese Alligator Reproduction, Xuancheng, Anhui 242000, China
| | - Liuyang Yang
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, Wuhu, Anhui 241000, China
| | - Huan Wang
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, Wuhu, Anhui 241000, China
| | - Yongkang Zhou
- Anhui Research Center of Chinese Alligator Reproduction, Xuancheng, Anhui 242000, China
| | - Genjun Tu
- Anhui Research Center of Chinese Alligator Reproduction, Xuancheng, Anhui 242000, China
| | - Yilin Shu
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
| | - Baowei Zhang
- School of Life Sciences, Anhui University, Hefei, Anhui 230601, China
| | - Xiaobing Wu
- College of Life Sciences, Anhui Normal University, Wuhu, Anhui 241000, China
- Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, Wuhu, Anhui 241000, China
| |
Collapse
|
32
|
Schulte L, Beck A, Marienfeld R, Azoitei N, Barth TFE, Beutel A, Benes V, Büchler MW, Gaisa NT, Kilani K, Giese N, Michalski CW, Möller P, Perkhofer L, Rausch T, Repky S, Roger E, Scheible J, Seufferlein T, Schirmacher P, Berger AW, Hackert T, Kleger A. Unveiling the intriguing relationship: oncogenic KRAS, morphological shifts, and mutational complexity in pancreatic mucinous cystic neoplasms. J Pathol 2025; 265:401-407. [PMID: 39906959 PMCID: PMC11880986 DOI: 10.1002/path.6397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 11/06/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) often arises from preexisting cystic lesions such as intraductal papillary mucinous neoplasms (IPMN) and mucinous cystic neoplasms (MCN). This study investigated the molecular heterogeneity and mutational landscape of MCN in relation to PDAC, highlighting the significance of KRAS mutations in tumor progression. Utilizing targeted next-generation sequencing on low-grade MCN and invasive PDAC samples, we identified a substantial overlap in mutational profiles, particularly mutations in KRAS, TP53, and FBXW7. Specifically, 69.2% of MCN exhibited somatic mutations, with KRAS mutations being a predominant oncogenic driver. The characterization of mutant versus wildtype KRAS variant allele frequencies (VAF) indicated higher mutation levels in PDAC compared to MCN, suggesting an evolutionary trajectory toward malignancy. Further histological analysis of 12 additional MCN cases revealed significant intratumor heterogeneity, with variant KRAS mutation distributions correlating with distinct cellular morphologies and dysplastic features. Additionally, we explored the potential of liquid biopsies, demonstrating a concordance rate of 71.4% for KRAS mutation detection in circulating tumor DNA (ctDNA) relative to tissue biopsies across cohorts. Our findings underscore the relevance of evaluating KRAS mutations-herein referred to as VAF per microdissected region-as they relate to histopathological markers of dysplasia, contributing to improved stratification of pancreatic lesions and facilitating personalized treatment strategies. In conclusion, this comprehensive analysis of MCN highlights the importance of KRAS as a crucial biomarker for both malignant progression and therapeutic decision-making in pancreatic pathology. Ultimately, our study suggests that characterizing the mutational landscape and histological features of MCN can enhance early detection and intervention strategies for at-risk patients. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
MESH Headings
- Humans
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/pathology
- Proto-Oncogene Proteins p21(ras)/genetics
- Mutation
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/pathology
- Male
- Female
- Biomarkers, Tumor/genetics
- Middle Aged
- Aged
- DNA Mutational Analysis
- High-Throughput Nucleotide Sequencing
- Neoplasms, Cystic, Mucinous, and Serous/genetics
- Neoplasms, Cystic, Mucinous, and Serous/pathology
- Genetic Predisposition to Disease
- Aged, 80 and over
Collapse
Affiliation(s)
- Lucas‐Alexander Schulte
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1Ulm University HospitalUlmGermany
| | - Annika Beck
- Department of PathologyUlm University HospitalUlmGermany
| | | | - Ninel Azoitei
- Institute of Molecular Oncology and Stem Cell Biology (IMOS)Ulm University HospitalUlmGermany
| | | | - Alica Beutel
- Department of Internal Medicine 1Ulm University HospitalUlmGermany
| | - Vladimir Benes
- Genomics Core FacilityEuropean Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Markus W Büchler
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | | | - Katja Kilani
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1Ulm University HospitalUlmGermany
| | - Nathalia Giese
- EPZ‐Biobank, Division of Pancreatic Cancer Research, Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Christoph W Michalski
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
| | - Peter Möller
- Department of PathologyUlm University HospitalUlmGermany
| | - Lukas Perkhofer
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1Ulm University HospitalUlmGermany
| | - Tobias Rausch
- Genomics Core FacilityEuropean Molecular Biology Laboratory (EMBL)HeidelbergGermany
| | - Stefan Repky
- Institute of Epidemiology and Medical BiometryUlm UniversityUlmGermany
| | - Elodie Roger
- Institute of Molecular Oncology and Stem Cell Biology (IMOS)Ulm University HospitalUlmGermany
| | - Jeanette Scheible
- Institute of Molecular Oncology and Stem Cell Biology (IMOS)Ulm University HospitalUlmGermany
| | | | - Peter Schirmacher
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Andreas Wolfgang Berger
- Department of Internal Medicine 1Ulm University HospitalUlmGermany
- Evangelisches Krankenhaus Königin Elisabeth HerzbergeDepartment of GastroenterologyBerlinGermany
| | - Thilo Hackert
- Department of General, Visceral and Transplantation SurgeryUniversity of HeidelbergHeidelbergGermany
- Department of General SurgeryUniversity Hospital Hamburg EppendorfHamburgGermany
| | - Alexander Kleger
- Division of Interdisciplinary Pancreatology, Department of Internal Medicine 1Ulm University HospitalUlmGermany
- Institute of Molecular Oncology and Stem Cell Biology (IMOS)Ulm University HospitalUlmGermany
| |
Collapse
|
33
|
Braun T, Rade M, Merz M, Klepzig H, Große F, Fandrei D, Pham NN, Kreuz M, Kuhn CK, Kuschel F, Löffler D, Meinel J, Heger E, Schweinsberg V, Pflug N, Platzbecker U, Hallek M, Holtick U, Köhl U, Scheid C, Reiche K, Herling M, Richardson T. Multiomic profiling of T cell lymphoma after therapy with anti-BCMA CAR T cells and GPRC5D-directed bispecific antibody. Nat Med 2025; 31:1145-1153. [PMID: 39984633 DOI: 10.1038/s41591-025-03499-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/07/2025] [Indexed: 02/23/2025]
Abstract
Chimeric antigen receptor (CAR) T cells and bispecific T cell engagers have become integral components in the treatment of relapsed/refractory multiple myeloma. We report a 63-year-old male who received ciltacabtagene autoleucel CAR T cells and the GPRC5D × CD3 bispecific talquetamab for early relapse of his multiple myeloma. Nine months after CAR T therapy, he developed a symptomatic leukemic peripheral T cell lymphoma with cutaneous and intestinal involvement. Longitudinal single-cell RNA and T cell receptor sequencing of peripheral blood and bone marrow revealed two hyperexpanded CAR-carrying T cell clones. These expanded clones exhibited an exhausted effector-memory T cell transcriptional signature, and the neoplasm itself was sensitive to dexamethasone treatment. The immunophenotypic and transcriptional alterations of these abnormal T cells resembled those of T-large granular lymphocytic leukemia. Spatial transcriptomes of skin lesions confirmed the aberrant CAR-expressing T cells. Whole-genome sequencing revealed three distinct integration sites, within the introns of ZGPAT, KPNA4 and polycomb-associated noncoding RNAs. Before and after CAR T whole-genome analyses implicated clonal outgrowth of a TET2-mutated precursor propelled by additional subclone-specific loss of heterozygosity and other secondary mechanisms. This case highlights the evolution of a CAR-carrying peripheral T cell lymphoma following CAR T cell and bispecific T cell engager therapy, offering critical insights into the clonal evolution from a predisposed hematopoietic precursor to a mature neoplasm.
Collapse
MESH Headings
- Humans
- Male
- Middle Aged
- Antibodies, Bispecific/therapeutic use
- Antibodies, Bispecific/immunology
- Receptors, G-Protein-Coupled/immunology
- Receptors, G-Protein-Coupled/genetics
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Multiple Myeloma/therapy
- Multiple Myeloma/immunology
- Multiple Myeloma/genetics
- Lymphoma, T-Cell/genetics
- Lymphoma, T-Cell/therapy
- Lymphoma, T-Cell/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Till Braun
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf (CIO ABCD), University Hospital Cologne, University of Cologne, Cologne, Germany
- Mildred Scheel School of Oncology Aachen Bonn Cologne Düsseldorf (MSSO ABCD), Cologne, Faculty of Medicine and University Hospital of Cologne, Cologne, Germany
| | - Michael Rade
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Maximilian Merz
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany.
- Department of Hematology, Cellular Therapy, Hemostaseology, Infectious Diseases, University Hospital of Leipzig, Leipzig, Germany.
- Cancer Center Central Germany (CCCG) Leipzig-Jena, University Hospital of Leipzig, Leipzig, Germany.
| | - Hanna Klepzig
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf (CIO ABCD), University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Florian Große
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Dresden/Leipzig, Germany
| | - David Fandrei
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Department of Hematology, Cellular Therapy, Hemostaseology, Infectious Diseases, University Hospital of Leipzig, Leipzig, Germany
- Cancer Center Central Germany (CCCG) Leipzig-Jena, University Hospital of Leipzig, Leipzig, Germany
| | - Nhu-Nguyen Pham
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Markus Kreuz
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | | | - Florian Kuschel
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf (CIO ABCD), University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Dennis Löffler
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Jörn Meinel
- Institute of Pathology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Eva Heger
- Institute of Virology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Viola Schweinsberg
- Department of Dermatology and Venereology, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Natali Pflug
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf (CIO ABCD), University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Hemostaseology, Infectious Diseases, University Hospital of Leipzig, Leipzig, Germany
- Cancer Center Central Germany (CCCG) Leipzig-Jena, University Hospital of Leipzig, Leipzig, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf (CIO ABCD), University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Udo Holtick
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf (CIO ABCD), University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ulrike Köhl
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Cancer Center Central Germany (CCCG) Leipzig-Jena, University Hospital of Leipzig, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Dresden/Leipzig, Germany
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Christof Scheid
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf (CIO ABCD), University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Kristin Reiche
- Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
- Cancer Center Central Germany (CCCG) Leipzig-Jena, University Hospital of Leipzig, Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Dresden/Leipzig, Germany
- Institute for Clinical Immunology, University Hospital of Leipzig, Leipzig, Germany
| | - Marco Herling
- Department of Hematology, Cellular Therapy, Hemostaseology, Infectious Diseases, University Hospital of Leipzig, Leipzig, Germany
- Cancer Center Central Germany (CCCG) Leipzig-Jena, University Hospital of Leipzig, Leipzig, Germany
| | - Tim Richardson
- Department I of Internal Medicine, Center for Integrated Oncology Aachen-Bonn-Cologne-Duesseldorf (CIO ABCD), University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
34
|
Perelli L, Zhang L, Mangiameli S, Giannese F, Mahadevan KK, Peng F, Citron F, Khan H, Le C, Gurreri E, Carbone F, Russell AJC, Soeung M, Lam TNA, Lundgren S, Marisetty S, Zhu C, Catania D, Mohamed AMT, Feng N, Augustine JJ, Sgambato A, Tortora G, Draetta GF, Tonon G, Futreal A, Giuliani V, Carugo A, Viale A, Kim MP, Heffernan TP, Wang L, Kalluri R, Cittaro D, Chen F, Genovese G. Evolutionary fingerprints of epithelial-to-mesenchymal transition. Nature 2025; 640:1083-1092. [PMID: 40044861 DOI: 10.1038/s41586-025-08671-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 01/17/2025] [Indexed: 04/13/2025]
Abstract
Mesenchymal plasticity has been extensively described in advanced epithelial cancers; however, its functional role in malignant progression is controversial1-5. The function of epithelial-to-mesenchymal transition (EMT) and cell plasticity in tumour heterogeneity and clonal evolution is poorly understood. Here we clarify the contribution of EMT to malignant progression in pancreatic cancer. We used somatic mosaic genome engineering technologies to trace and ablate malignant mesenchymal lineages along the EMT continuum. The experimental evidence clarifies the essential contribution of mesenchymal lineages to pancreatic cancer evolution. Spatial genomic analysis, single-cell transcriptomic and epigenomic profiling of EMT clarifies its contribution to the emergence of genomic instability, including events of chromothripsis. Genetic ablation of mesenchymal lineages robustly abolished these mutational processes and evolutionary patterns, as confirmed by cross-species analysis of pancreatic and other human solid tumours. Mechanistically, we identified that malignant cells with mesenchymal features display increased chromatin accessibility, particularly in the pericentromeric and centromeric regions, in turn resulting in delayed mitosis and catastrophic cell division. Thus, EMT favours the emergence of genomic-unstable, highly fit tumour cells, which strongly supports the concept of cell-state-restricted patterns of evolution, whereby cancer cell speciation is propagated to progeny within restricted functional compartments. Restraining the evolutionary routes through ablation of clones capable of mesenchymal plasticity, and extinction of the derived lineages, halts the malignant potential of one of the most aggressive forms of human cancer.
Collapse
Affiliation(s)
- Luigi Perelli
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Li Zhang
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah Mangiameli
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | | | - Krishnan K Mahadevan
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fuduan Peng
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francesca Citron
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hania Khan
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney Le
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enrico Gurreri
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | | | - Andrew J C Russell
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Melinda Soeung
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Truong Nguyen Anh Lam
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sebastian Lundgren
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sujay Marisetty
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cihui Zhu
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Desiree Catania
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alaa M T Mohamed
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ningping Feng
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jithesh Jose Augustine
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Multiplex Spatial Imaging Facility, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giampaolo Tortora
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giulio F Draetta
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Giovanni Tonon
- Center for Omics Sciences, IRCCS San Raffaele Institute, Milan, Italy
| | - Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Virginia Giuliani
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Andrea Viale
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael P Kim
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy P Heffernan
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center, UT Health Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, USA
| | - Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Davide Cittaro
- Center for Omics Sciences, IRCCS San Raffaele Institute, Milan, Italy.
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| | - Giannicola Genovese
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- TRACTION Platform, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
35
|
Jiao D, Dong X, Fan S, Liu X, Yu Y, Wei C. Gastric cancer genomics study using reference human pangenomes. Life Sci Alliance 2025; 8:e202402977. [PMID: 39870503 PMCID: PMC11772497 DOI: 10.26508/lsa.202402977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/29/2025] Open
Abstract
A pangenome is the sum of the genetic information of all individuals in a species or a population. Genomics research has been gradually shifted to a paradigm using a pangenome as the reference. However, in disease genomics study, pangenome-based analysis is still in its infancy. In this study, we introduced a graph-based pangenome GGCPan from 185 patients with gastric cancer. We then systematically compared the cancer genomics study results using GGCPan, a linear pangenome GCPan, and the human reference genome as the reference. For small variant detection and microsatellite instability status identification, there is little difference in using three different genomes. Using GGCPan as the reference had a significant advantage in structural variant identification. A total of 24 candidate gastric cancer driver genes were detected using three different reference genomes, of which eight were common and five were detected only based on pangenomes. Our results showed that disease-specific pangenome as a reference is promising and a whole set of tools are still to be developed or improved for disease genomics study in the pangenome era.
Collapse
Affiliation(s)
- Du Jiao
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaorui Dong
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyu Fan
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyi Liu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaochun Wei
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
36
|
Delaive S, Sylvestre F, Xuereb A, Lecomte L, Boyle B, Otis C, Bernatchez L, Derome N. Population Genetic Structure of Three-Spined Sticklebacks in the St. Lawrence: A Gradient of Change. Ecol Evol 2025; 15:e71153. [PMID: 40270791 PMCID: PMC12015749 DOI: 10.1002/ece3.71153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 04/25/2025] Open
Abstract
Understanding how environmental gradients shape population genetic structure is critical for elucidating evolutionary dynamics in heterogeneous landscapes. The St. Lawrence Estuary, spanning fluvial, middle, and marine zones, presents a steep salinity gradient that serves as an ideal setting to study such a question. Three-spined sticklebacks (Gasterosteus aculeatus) thrive across these zones, offering an ideal model system to investigate the interplay of gene flow and natural selection in shaping population structure. Using whole-genome resequencing of sticklebacks from 12 sites, this study aimed to resolve fine-scale population structure and investigate how genetic diversity and differentiation are influenced by selection and gene flow. By integrating single nucleotide polymorphisms (SNPs) and structural variants (SVs), we assessed differentiation patterns, examined clinal variation, and evaluated the relative roles of gene flow and selection in shaping population dynamics. Our findings reveal clear genetic differentiation between fluvial and saltwater populations, with Baie-Saint-Paul forming a potential third group. Salinity emerged as a key driver of genetic structure, with clinal variation in allele frequencies suggesting ongoing adaptation along the gradient. Demographic modeling indicated a history of secondary contact with recent and weak gene flow. Structural variants, particularly indels, complemented SNP-based analyses, underscoring their importance in detecting fine-scale population structure. These results highlight the complex interplay of evolutionary forces shaping biodiversity in transitional environments, providing a basis for exploring local adaptation in connected populations and contributing to broader efforts in conservation genomics.
Collapse
Affiliation(s)
- Sann Delaive
- Département de Biologie, Institut de Biologie Intégrative et Des Systèmes (IBIS)Université LavalQuébecQuebecCanada
| | - Florent Sylvestre
- Département de Biologie, Institut de Biologie Intégrative et Des Systèmes (IBIS)Université LavalQuébecQuebecCanada
| | - Amanda Xuereb
- Département de Biologie, Institut de Biologie Intégrative et Des Systèmes (IBIS)Université LavalQuébecQuebecCanada
| | - Laurie Lecomte
- Département de Biologie, Institut de Biologie Intégrative et Des Systèmes (IBIS)Université LavalQuébecQuebecCanada
| | - Brian Boyle
- Institut de Biologie Intégrative et Des SystèmesUniversité LavalQuébecQuebecCanada
| | - Christian Otis
- Institut de Biologie Intégrative et Des SystèmesUniversité LavalQuébecQuebecCanada
| | | | - Nicolas Derome
- Département de Biologie, Institut de Biologie Intégrative et Des Systèmes (IBIS)Université LavalQuébecQuebecCanada
| |
Collapse
|
37
|
Akarca FG, Grenert JP, Kakar S. Role of genomic analysis in the classification of well differentiated hepatocellular lesions. Hum Pathol 2025; 158:105794. [PMID: 40374146 DOI: 10.1016/j.humpath.2025.105794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND The distinction of focal nodular hyperplasia (FNH) and hepatocellular adenoma (HCA) from well-differentiated hepatocellular carcinoma (WD-HCC) in noncirrhotic liver can be challenging. High-grade dysplastic nodule (HGDN) in cirrhosis can have overlapping features with WD-HCC. In some cases, HCA diagnosis is evident but glutamine synthetase (GS) staining is indeterminate for β-catenin activation, which does not allow reliable risk assessment. This study examines the role of genomic analysis in better categorization of WD hepatocellular lesions (WDHL). DESIGN Genomic analysis using capture-based NGS assay was done in 23 WDHLs that could not be definitely classified based on morphology, reticulin stain and IHC, and were designated as 'atypical hepatocellular neoplasms' (AHNs). GS staining was classified as diffuse homogeneous (moderate to strong staining in >90 % of tumor cells), diffuse heterogeneous (50-90 %), not diffuse (<50 %) and borderline (not clear if more or less than 50 %). RESULTS The genomic profile provided additional information for the diagnosis and/or risk assessment enabling a benign diagnosis in 15/23 cases (66 %) and HCC in 4/23 cases (17 %), while the diagnosis remained as atypical in the remaining 4 cases. Of the 4 cases with final HCC diagnosis, findings were suspicious but not diagnostic based on morphology/IHC; additional changes like TERT promoter mutation (n = 2), AXIN mutation (n = 1), CDKN2A loss (n = 2) and copy number alterations (n = 3) helped to support HCC. Of the 15 cases with a final benign diagnosis, the status of β-catenin activation was unclear based on GS stain in 8 cases, 2 of which showed CTNNB1 exon 7 mutation, 1 showed CTNNB1 exon 8 mutation, while genomic changes in 5 cases did not show any evidence of Wnt activation. FNH-like features were seen in 2 cases, but the genomic changes excluded FNH (CTNNB1 and ARID1A mutation). The final diagnosis was unchanged from the initial diagnosis of AHN in 4/23 cases (17 %) as the molecular findings did not favor HCC. CONCLUSION Genomic changes were helpful in characterization of WDHLs, supporting HCC in 17 % of cases and clarifying β-catenin activation status in all 7 cases with borderline GS staining. Genomic changes are not specific but can provide diagnostic clues in selected challenging cases that cannot be classified on morphology and IHC. Given the significant treatment implications of distinguishing between HCC and benign/premalignant entities, routine use of genomic analysis in diagnostically challenging settings should be considered.
Collapse
MESH Headings
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/classification
- Liver Neoplasms/diagnosis
- Liver Neoplasms/chemistry
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/classification
- Carcinoma, Hepatocellular/chemistry
- Carcinoma, Hepatocellular/diagnosis
- Adenoma, Liver Cell/genetics
- Adenoma, Liver Cell/pathology
- Adenoma, Liver Cell/classification
- Adenoma, Liver Cell/diagnosis
- Adenoma, Liver Cell/chemistry
- Male
- Middle Aged
- Female
- Aged
- Focal Nodular Hyperplasia/genetics
- Focal Nodular Hyperplasia/pathology
- Focal Nodular Hyperplasia/diagnosis
- Focal Nodular Hyperplasia/classification
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/analysis
- Adult
- Genomics/methods
- Diagnosis, Differential
- Glutamate-Ammonia Ligase/analysis
- beta Catenin/genetics
- Mutation
- High-Throughput Nucleotide Sequencing
- Predictive Value of Tests
- Cell Differentiation
Collapse
|
38
|
Wang Z, Pan D, Xie X, Zhong Z, Wang F, Xiao Q. Genome-wide detection of runs of homozygosity in Ding'an pigs revealed candidate genes relating to meat quality traits. BMC Genomics 2025; 26:316. [PMID: 40165050 PMCID: PMC11956453 DOI: 10.1186/s12864-025-11501-4] [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: 12/24/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Ding'an (DA) pig, a native Chinese breed, is renowned for its excellent meat quality, disease resistance, high reproductive performance, and adaptability. Its meat quality traits hold significant economic value. However, its conservation population has been declining due to the impact of commercialized breeds and African swine fever, which is not conducive to its development and utilization. RESULTS This study utilized whole-genome resequencing data from 15 DA pigs to reveal their genetic characteristics and current resource status. We analyzed the length, number, and distribution patterns of Runs of Homozygosity (ROH) in DA pigs, as well as high-frequency ROH regions. The results identified 23,208,098 single nucleotide polymorphisms (SNPs), 4,497,242 insertion and deletion (InDels), 13,622 copy number variation (CNVs), and 399,934 structure variation (SVs). Further analysis revealed relatively high genetic diversity and low inbreeding levels in DA pigs. Through functional gene enrichment analysis of high-frequency ROH regions, we identified multiple candidate genes associated with specific traits in DA pigs, including meat quality (ANKRD1, CPNE5, MYOM1), fat deposition (OBSCN, MAPK4, PNPLA1, PACSIN1, GRM4), and skeletal muscle development (LRPPRC, WNT9A). CONCLUSIONS This study conducted whole-genome sequencing and ROH analysis on DA pigs, revealing high genetic diversity and low inbreeding levels within the population. Through functional gene enrichment analysis of high-frequency ROH regions, we identified multiple candidate genes associated with meat quality, fat deposition, and skeletal muscle development. These findings not only enhance our understanding of the genetic mechanisms underlying the unique traits of DA pigs but also provide valuable insights for practical applications. Specifically, the identified candidate genes and genomic regions can guide conservation efforts to maintain genetic diversity and mitigate inbreeding risks. Meanwhile, these genetic insights can be integrated into breeding programs to improve meat quality and other economically important traits, thereby supporting the sustainable development and utilization of DA pigs.
Collapse
Affiliation(s)
- Ziyi Wang
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou, 570228, China
| | - Deyou Pan
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou, 570228, China
| | - Xinfeng Xie
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou, 570228, China
| | - Ziqi Zhong
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou, 570228, China
| | - Feng Wang
- Institute of Animal Science and Veterinary Medicine, Hainan Academy of Agricultural Sciences, Haikou, 571100, China
| | - Qian Xiao
- Hainan Key Laboratory of Tropical Animal Reproduction & Breeding and Epidemic Disease Research, School of Tropical Agriculture and Forestry, Hainan University, Haikou, 570228, China.
| |
Collapse
|
39
|
Guo Q, Li Y, Wang TY, Ramakrishnan A, Yang R. OctopusV and TentacleSV: a one-stop toolkit for multi-sample, cross-platform structural variant comparison and analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645012. [PMID: 40196604 PMCID: PMC11974888 DOI: 10.1101/2025.03.24.645012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Structural variants (SVs) significantly influence genomic variability and disease, but their accurate analysis across multiple samples and sequencing platforms remains challenging. We developed OctopusV, a tool that standardizes ambiguous breakend (BND) annotations into canonical SV types (inversions, duplications, translocations) and integrates variant calls using flexible set operations, such as union, intersection, difference, and complement, enabling cohort-specific variant identification. Together with TentacleSV, an automated pipeline, OctopusV provides an end-to-end solution from raw data to final callsets. Evaluations show improved precision, recall, and consistency, highlighting its value in cancer genomics and rare disease diagnostics. Both tools are available at https://github.com/ylab-hi/OctopusV and https://github.com/ylab-hi/TentacleSV.
Collapse
Affiliation(s)
- Qingxiang Guo
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E Superior St, Chicago, 60611, IL, USA
| | - Yangyang Li
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E Superior St, Chicago, 60611, IL, USA
| | - Ting-You Wang
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E Superior St, Chicago, 60611, IL, USA
| | - Abhi Ramakrishnan
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E Superior St, Chicago, 60611, IL, USA
| | - Rendong Yang
- Department of Urology, Northwestern University Feinberg School of Medicine, 303 E Superior St, Chicago, 60611, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, 675 N St Clair St, Chicago, 60611, IL, USA
| |
Collapse
|
40
|
Berghöfer J, Khaveh N, Mundlos S, Metzger J. Multi-tool copy number detection highlights common body size-associated variants in miniature pig breeds from different geographical regions. BMC Genomics 2025; 26:285. [PMID: 40121435 PMCID: PMC11929999 DOI: 10.1186/s12864-025-11446-8] [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: 10/30/2024] [Accepted: 03/05/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Copy number variations (CNVs) represent a common and highly specific type of variation in the genome, potentially influencing genetic diversity and mammalian phenotypic development. Structural variants, such as deletions, duplications, and insertions, have frequently been highlighted as key factors influencing traits in high-production pigs. However, comprehensive CNV analyses in miniature pig breeds are limited despite their value in biomedical research. RESULTS This study performed whole-genome sequencing in 36 miniature pigs from nine breeds from America, Asia and Oceania, and Europe. By employing a multi-tool approach (CNVpytor, Delly, GATK gCNV, Smoove), the accuracy of CNV identification was improved. In total, 34 homozygous CNVs overlapped with exonic regions in all samples, suggesting a role in expressing specific phenotypes such as uniform growth patterns, fertility, or metabolic function. In addition, 386 copy number variation regions (CNVRs) shared by all breeds were detected, covering 33.6 Mb (1.48% of the autosomal genome). Further, 132 exclusive CNVRs were identified for American breeds, 47 for Asian and Oceanian breeds, and 114 for European breeds. Functional enrichment analysis revealed genes within the common CNVRs involved in body height determination and other growth-related parameters. Exclusive CNVRs were located in the region of genes enriched for lipid metabolism in American minipigs, reproductive traits in Asian and Oceanian breeds, and cardiovascular features and body height in European breeds. In the selected groups, quantitative trait loci associated with body size, meat quality, reproduction, and disease susceptibility were highlighted. CONCLUSION This investigation of the CNV landscape of minipigs underlines the impact of selective breeding on structural variants and its role in the development of specific breed phenotypes across geographical areas. The multi-tool approach provides a valuable resource for future studies on the effects of artificial selection on livestock genomes.
Collapse
Affiliation(s)
- Jan Berghöfer
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
- Institute of Animal Genomics, University of Veterinary Medicine Hanover, Hanover, Germany
| | - Nadia Khaveh
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Animal Genomics, University of Veterinary Medicine Hanover, Hanover, Germany
| | - Stefan Mundlos
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute for Medical and Human Genetics, Charité Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, BCRT - Berlin Institute of Health Centre for Regenerative Therapies, Berlin, Germany
| | - Julia Metzger
- Max Planck Institute for Molecular Genetics, Berlin, Germany.
- Institute of Animal Genomics, University of Veterinary Medicine Hanover, Hanover, Germany.
| |
Collapse
|
41
|
Chen A, Zhao X, Wang H, Zhao X, Wang G, Zhang X, Ren X, Zhang Y, Cheng X, Yu X, Mei X, Wang H, Guo M, Jiang X, Zhang F, Ning Z, Qu L. Genetic parameter estimation and molecular foundation of Double-yolk eggs trait in white leghorn. Poult Sci 2025; 104:105069. [PMID: 40300322 PMCID: PMC12059378 DOI: 10.1016/j.psj.2025.105069] [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: 01/03/2025] [Revised: 03/06/2025] [Accepted: 03/18/2025] [Indexed: 05/01/2025] Open
Abstract
Double-yolk (DY) eggs refer to the presence of two yolks in an egg, and they are often present in fowl flocks. As abnormal eggs, DY eggs occur frequently in the early stages of egg-laying in hens, as well as in hens with early sexual maturity. In order to understand the inheritant pattern of DY eggs and explore candidate genes associated with DY eggs, we selected over 10,000 white Leghorn (WL) chickens from 4 generations and recorded the data during the egg laying period, including total egg number and the rate of DY egg number during the first 2 months and the period of 18-58 weeks of age (EN2month, EN38, EN58, DY2month, DY38, and DY58), age at first egg (AFE), first egg weight (FEW), and body weight at first egg (BWA). The results of genetic parameter analysis showed that the DY egg rate was a trait with low to medium heritabilities with the values from 0.15 to 0.29. And there were strong positive phenotypic and genetic correlations between DY egg rate and egg production at different age stages, and they were all strongly negatively correlated with AFE. However, the DY egg rate and egg production at different stages had strong positive and negative genetic correlations with BWA and FEW, respectively. We also found that significant differences in these trait values between different generations and cage layers, indicating that generations and cage layers had a certain influence on these traits. Furtherly, we used whole genome-wide association (GWA) analysis to identify genes underlying DY, and 5 candidate genes (EZH2, CNTNAP2, TMEM163, GPC1, and ACMSD) associated with DY2month in WL. Our study improved the understanding of DY eggs in hens, and the genetic parameters of DY eggs, and also provided insights into reducing the production of DY eggs by various selection strategies.
Collapse
Affiliation(s)
- Anqi Chen
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xiaoyu Zhao
- Xingrui Agricultural Stock Breeding, Baoding 072550, Hebei Province, PR China
| | - Haiyan Wang
- Hohhot Customs District, Hohhot 010010, Inner Mongolia Autonomous Region, PR China
| | - Xiurong Zhao
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Gang Wang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xinye Zhang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xufang Ren
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Yalan Zhang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xue Cheng
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xiaofan Yu
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xiaohan Mei
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Huie Wang
- Xinjiang Production and Construction Corps, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, Tarim University, Alar 843300, PR China
| | - Menghan Guo
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xiaoyu Jiang
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Fuping Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Sciences, Guizhou University, Guiyang 550025, Guizhou Province, PR China
| | - Zhonghua Ning
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Lujiang Qu
- National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China; Xinjiang Production and Construction Corps, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, Tarim University, Alar 843300, PR China.
| |
Collapse
|
42
|
Shaver AO, McKeown R, Otero JMR, Andersen EC. Independent mechanisms of benzimidazole resistance across Caenorhabditis nematodes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643047. [PMID: 40161721 PMCID: PMC11952494 DOI: 10.1101/2025.03.13.643047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Benzimidazoles (BZs), a widely used class of anthelmintic drugs, target beta-tubulin proteins, disrupt microtubule formation, and cause nematode death. In parasitic nematode species, mutations in beta-tubulin genes (e.g., isotype-1 beta-tubulin) are predicted to inhibit BZ binding and are associated with BZ resistance. Similarly, in the free-living nematode Caenorhabditis elegans, mutations in an isotype-1 beta-tubulin ortholog, ben-1, are the primary drivers of BZ resistance. The recurrent association of BZ resistance with beta-tubulins suggests that BZ resistance is repeatedly caused by mutations in beta-tubulin genes, an example of repeated evolution of drug resistance across nematode species. To evaluate the hypothesis of repeated evolution of BZ resistance mediated by beta-tubulin, we identified predicted resistance alleles in beta-tubulin genes across wild strains from three Caenorhabditis species: C. elegans, Caenorhabditis briggsae, and Caenorhabditis tropicalis. We hypothesized that, if these species experienced similar selective pressures, they would evolve resistance to BZs by mutations in any of three beta-tubulin genes (ben-1, tbb-1, and tbb-2). Using high-throughput development assays, we tested the association of predicted beta-tubulin alleles with BZ resistance. We found that a heterogeneous set of variants identified in C. elegans ben-1 were associated with BZ resistance. In C. briggsae, only two variants in ben-1, predicted to encode a premature stop codon (W21stop) and a missense substitution (Q134H), were associated with BZ resistance. In C. tropicalis, two missense variants were identified in ben-1, but neither was associated with BZ resistance. C. briggsae and C. tropicalis might have evolved BZ resistance by mutations in other beta-tubulin genes, but we found that variants in tbb-1 or tbb-2 in these species were not associated with BZ resistance. Our findings reveal a lack of repeated evolution of BZ resistance across the three Caenorhabditis species and highlight the importance of defining BZ resistance mechanisms outside of beta-tubulins.
Collapse
Affiliation(s)
- Amanda O. Shaver
- Dept. of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ryan McKeown
- Dept. of Molecular Biosciences, Northwestern University, Evanston, Illinois, USA
| | | | - Erik C. Andersen
- Dept. of Biology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
43
|
Verschuren J, van Schendel R, van Bostelen I, Verkennis AEE, Knipscheer P, Tijsterman M. FAN1-mediated translesion synthesis and POLQ/HELQ-mediated end joining generate interstrand crosslink-induced mutations. Nat Commun 2025; 16:2495. [PMID: 40082407 PMCID: PMC11906846 DOI: 10.1038/s41467-025-57764-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: 01/25/2024] [Accepted: 03/04/2025] [Indexed: 03/16/2025] Open
Abstract
To counteract the damaging effects of DNA interstrand crosslinks (ICLs), cells have evolved various specialized ICL repair pathways. However, how ICL repair impacts genetic integrity remains incompletely understood. Here, we determined the mutagenic consequences of psoralen ICL repair in the animal model C. elegans and identify two mutagenic repair mechanisms: (i) translesion synthesis through POLH and REV1/3-mediated bypass, leading to single nucleotide polymorphisms (SNVs), and (ii) end joining via POLQ or HELQ action resulting in deletions. While we found no role for the Fanconi anemia genes FANCD2 and FANCI, disruption of TRAIP, which triggers unloading of the CMG helicase at sites of blocked replication, led to a strikingly altered repair profile, suggesting a role for DNA replication in the etiology of ICL-induced deletions. TRAIP deficiency did not affect SNV formation; instead, we found these SNVs to depend on the functionality of the Fanconi anemia-associated nuclease FAN1.
Collapse
Affiliation(s)
- Jip Verschuren
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Robin van Schendel
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ivo van Bostelen
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Alex E E Verkennis
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Puck Knipscheer
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Marcel Tijsterman
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
44
|
Aydin SK, Yilmaz KC, Acar A. Benchmarking long-read structural variant calling tools and combinations for detecting somatic variants in cancer genomes. Sci Rep 2025; 15:8707. [PMID: 40082509 PMCID: PMC11906795 DOI: 10.1038/s41598-025-92750-x] [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: 10/24/2024] [Accepted: 03/03/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer genomes have a complicated landscape of mutations, including large-scale rearrangements known as structural variants (SVs). These SVs can disrupt genes or regulatory elements, playing a critical role in cancer development and progression. Despite their importance, accurate identification of somatic structural variants (SVs) remains a significant bottleneck in cancer genomics. Long-read sequencing technologies hold great promise in SV discovery, and there is an increasing number of efforts to develop new tools to detect them. In this study, we employ eight widely used SV callers on paired tumor and matched normal samples from both the NCI-H2009 lung cancer cell line and the COLO829 melanoma cell line, the latter of which has a well-established somatic SV truth set. Following separate variation detection in both tumor and normal DNA, the VCF merging procedure and a subtraction method were used to identify candidate somatic SVs. Additionally, we explored different combinations of the tools to enhance the accuracy of true somatic SV detection. Our analysis adopts a comprehensive approach, evaluating the performance of each SV caller across a spectrum of variant types and numbers in finding cancer-related somatic SVs. This study, by comparing eight different tools and their combinations, not only reveals the benefits and limitations of various techniques but also establishes a framework for developing more robust SV calling pipelines. Our findings highlight the strengths and weaknesses of current SV calling tools and suggest that combining multiple tools and testing different combinations can significantly enhance the validation of somatic alterations.
Collapse
Affiliation(s)
- Safa Kerem Aydin
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupınar Bulvarı 1, 06800, Çankaya, Ankara, Turkey
| | - Kubra Celikbas Yilmaz
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupınar Bulvarı 1, 06800, Çankaya, Ankara, Turkey
| | - Ahmet Acar
- Department of Biological Sciences, Middle East Technical University, Universiteler Mah. Dumlupınar Bulvarı 1, 06800, Çankaya, Ankara, Turkey.
| |
Collapse
|
45
|
Huyghebaert J, Christiaenssen B, De Rademaeker M, Van den Ende J, Vandeweyer G, Kooy RF, Mateiu L, Annear D. Paracentric inversion disrupting the SHANK2 gene. Eur J Med Genet 2025; 75:105009. [PMID: 40057302 DOI: 10.1016/j.ejmg.2025.105009] [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: 08/30/2024] [Revised: 02/07/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
In this study, we employed a multifaceted approach combining short-read whole genome sequencing (WGS) analyzed using Delly, cytogenomics using Bionano technology, and Sanger sequencing to identify the breakpoints of a balanced de novo paracentric inversion on chromosome 11, spanning approximately 64 Mb (inv11q13.3; q25). This inversion was discovered in a girl who presented with mild intellectual disability (ID), speech and language delays, a delay in motor development and attention deficit hyperactivity disorder (ADHD). Detailed analysis of the breakpoints revealed the disruption of two genes; SHANK2, which is critical for encoding a postsynaptic scaffolding protein at glutamatergic synapses in the brain, and LINC02714, a long non-coding RNA (lncRNA). Although SHANK2 is not listed in the OMIM database as a causative gene to this date, literature reports at least 21 cases where (likely) pathogenic variants in SHANK2 have been identified in patients with neurodevelopmental disorders (NDDs). A loss of function variant of the SHANK2 gene is in line with the clinical presentation of this patient. No additional genetic variants that could explain her phenotype were identified. In conclusion, by combining WGS, cytogenomics and Sanger sequencing techniques, we identified the exact breakpoints of a large inversion providing a likely molecular diagnosis for our patient.
Collapse
Affiliation(s)
- Jolien Huyghebaert
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium.
| | | | - Marjan De Rademaeker
- Department of Medical Genetics, University Hospital of Antwerp, Antwerp, Belgium.
| | - Jenneke Van den Ende
- Department of Medical Genetics, University Hospital of Antwerp, Antwerp, Belgium.
| | - Geert Vandeweyer
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium; Department of Medical Genetics, University Hospital of Antwerp, Antwerp, Belgium.
| | - R Frank Kooy
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium.
| | - Ligia Mateiu
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium.
| | - Dale Annear
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium; Department of Biochemistry & Microbiology, Nelson Mandela University, Gqeberha, South Africa.
| |
Collapse
|
46
|
Zhao S, Xu D, Cai J, Shen Q, He M, Pan X, Gao Y, Li J, Yuan X. Benchmarking strategies for CNV calling from whole genome bisulfite data in humans. Comput Struct Biotechnol J 2025; 27:912-919. [PMID: 40123798 PMCID: PMC11929052 DOI: 10.1016/j.csbj.2025.02.040] [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] [Indexed: 03/25/2025] Open
Abstract
It's important to dissect the relationship between copy number variations (CNVs) and DNA methylation, because both greatly change the dosages of genes and are responsible for diverse human cancers. Although whole genome bisulfite sequencing (WGBS) informs CNVs and DNA methylation, no study has provided a systematic benchmark for detecting CNVs from WGBS data. Herein, based on simulated and real WGBS datasets of 84.62 billion reads, we undertook 714 CNV detections to comprehensively benchmark the performance of 35 strategies, 5 alignment algorithms (bismarkbt2, bsbolt, bsmap, bwameth, and walt) wrapping with 7 CNV detection applications (BreakDancer, cn.mops, CNVkit, CNVnator, DELLY, GASV and Pindel). The results highlighted a subset of strategies that accurately called CNVs depending on numbers, lengths, precision, recall, and F1 scores of CNV detections. We found that bwameth-DELLY and bwameth-BreakDancer were the best strategies for calling deletions, and walt-CNVnator and bismarkbt2-CNVnator were the best strategies for calling duplications. These works provided investigators with useful information to accurately explore CNVs from WGBS data in humans.
Collapse
Affiliation(s)
- Shanghui Zhao
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Dantong Xu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jiali Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Qingpeng Shen
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Mingran He
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Xiangchun Pan
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Yahui Gao
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jiaqi Li
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Xiaolong Yuan
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, Guangdong Provincial Key Laboratory of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
- National Center of Technology Innovation for Pigs, Chongqing 402460, China
- Centre for Healthy Ageing, Health Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| |
Collapse
|
47
|
Mouton A, Bird DJ, Li G, Craven BA, Levine JM, Morselli M, Pellegrini M, Van Valkenburgh B, Wayne RK, Murphy WJ. Genetic and Anatomical Determinants of Olfaction in Dogs and Wild Canids. Mol Biol Evol 2025; 42:msaf035. [PMID: 39950968 PMCID: PMC11950533 DOI: 10.1093/molbev/msaf035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 03/29/2025] Open
Abstract
Understanding the anatomical and genetic basis of complex phenotypic traits has long been a challenge for biological research. Domestic dogs offer a compelling model as they demonstrate more phenotypic variation than any other vertebrate species. Dogs have been intensely selected for specific traits and abilities, directly or indirectly, over the past 15,000 years since their initial domestication from the gray wolf. Because olfaction plays a central role in critical tasks, such as the detection of drugs, diseases, and explosives, as well as human rescue, we compared relative olfactory capacity across dog breeds and assessed changes to the canine olfactory system to their direct ancestors, wolves, and coyotes. We conducted a cross-disciplinary survey of olfactory anatomy, olfactory receptor (OR) gene variation, and OR gene expression in domestic dogs. Through comparisons to their closest wild canid relatives, the gray wolf and coyote, we show that domestic dogs might have lost functional OR genes commensurate with a documented reduction in nasal morphology as an outcome of the domestication process prior to breed formation. Critically, within domestic dogs alone, we found no genetic or morphological profile shared among functional or genealogical breed groupings, such as scent hounds, that might indicate evidence of any human-directed selection for enhanced olfaction. Instead, our results suggest that superior scent detection dogs likely owe their success to advantageous behavioral traits and training rather than an "olfactory edge" provided by morphology or genes.
Collapse
Affiliation(s)
- Alice Mouton
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, USA
- InBios-Conservation Genetics Lab, University of Liege, Liège, Belgium
| | - Deborah J Bird
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, USA
| | - Gang Li
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
- College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Brent A Craven
- Department of Mechanical and Nuclear Engineering, Pennsylvania State University, University Park, PA, USA
| | - Jonathan M Levine
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Marco Morselli
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, CA, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, CA, USA
| | - Blaire Van Valkenburgh
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, USA
| | - Robert K Wayne
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, USA
| | - William J Murphy
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
48
|
Vialle RA, de Paiva Lopes K, Li Y, Ng B, Schneider JA, Buchman AS, Wang Y, Farfel JM, Barnes LL, Wingo AP, Wingo TS, Seyfried NT, De Jager PL, Gaiteri C, Tasaki S, Bennett DA. Structural variants linked to Alzheimer's disease and other common age-related clinical and neuropathologic traits. Genome Med 2025; 17:20. [PMID: 40038788 PMCID: PMC11881306 DOI: 10.1186/s13073-025-01444-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a complex neurodegenerative disorder with substantial genetic influence. While genome-wide association studies (GWAS) have identified numerous risk loci for late-onset AD (LOAD), the functional mechanisms underlying most of these associations remain unresolved. Large genomic rearrangements, known as structural variants (SVs), represent a promising avenue for elucidating such mechanisms within some of these loci. METHODS By leveraging data from two ongoing cohort studies of aging and dementia, the Religious Orders Study and Rush Memory and Aging Project (ROS/MAP), we performed genome-wide association analysis testing 20,205 common SVs from 1088 participants with whole genome sequencing (WGS) data. A range of Alzheimer's disease and other common age-related clinical and neuropathologic traits were examined. RESULTS First, we mapped SVs across 81 AD risk loci and discovered 22 SVs in linkage disequilibrium (LD) with GWAS lead variants and directly associated with the phenotypes tested. The strongest association was a deletion of an Alu element in the 3'UTR of the TMEM106B gene, in high LD with the respective AD GWAS locus and associated with multiple AD and AD-related disorders (ADRD) phenotypes, including tangles density, TDP-43, and cognitive resilience. The deletion of this element was also linked to lower TMEM106B protein abundance. We also found a 22-kb deletion associated with depression in ROS/MAP and bearing similar association patterns as GWAS SNPs at the IQCK locus. In addition, we leveraged our catalog of SV-GWAS to replicate and characterize independent findings in SV-based GWAS for AD and five other neurodegenerative diseases. Among these findings, we highlight the replication of genome-wide significant SVs for progressive supranuclear palsy (PSP), including markers for the 17q21.31 MAPT locus inversion and a 1483-bp deletion at the CYP2A13 locus, along with other suggestive associations, such as a 994-bp duplication in the LMNTD1 locus, suggestively linked to AD and a 3958-bp deletion at the DOCK5 locus linked to Lewy body disease (LBD) (P = 3.36 × 10-4). CONCLUSIONS While still limited in sample size, this study highlights the utility of including analysis of SVs for elucidating mechanisms underlying GWAS loci and provides a valuable resource for the characterization of the effects of SVs in neurodegenerative disease pathogenesis.
Collapse
Affiliation(s)
- Ricardo A Vialle
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA.
| | - Katia de Paiva Lopes
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Yan Li
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Bernard Ng
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Julie A Schneider
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Aron S Buchman
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Yanling Wang
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Jose M Farfel
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - Aliza P Wingo
- Department of Psychiatry, University of California, Davis, Davis, CA, USA
- VA Northern California Health Care System, Davis, CA, USA
| | - Thomas S Wingo
- Department of Neurology, University of California, Davis, Davis, CA, USA
| | - Nicholas T Seyfried
- Department of Neurology and Department of Biochemistry, Goizueta Alzheimer's Disease Research Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Chris Gaiteri
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Shinya Tasaki
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, 1750 W Harrison St, Chicago, IL, 60612, USA
| |
Collapse
|
49
|
Xiao H, Wang Y, Liu W, Shi X, Huang S, Cao S, Long Q, Wang X, Liu Z, Xu X, Peng Y, Wang P, Jiang Z, Riaz S, Walker AM, Gaut BS, Huang S, Zhou Y. Impacts of reproductive systems on grapevine genome and breeding. Nat Commun 2025; 16:2031. [PMID: 40032836 PMCID: PMC11876636 DOI: 10.1038/s41467-025-56817-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/31/2025] [Indexed: 03/05/2025] Open
Abstract
Diversified reproductive systems can be observed in the plant kingdom and applied in crop breeding; however, their impacts on crop genomic variation and breeding remain unclear. Grapevine (Vitis vinifera L.), a widely planted fruit tree, underwent a shift from dioecism to monoecism during domestication and involves crossing, self-pollination, and clonal propagation for its cultivation. In this study, we discover that the reproductive types, namely, crossing, selfing, and cloning, dramatically impact genomic landscapes and grapevine breeding based on comparative genomic and population genetics of wild grapevine and a complex pedigree of Pinot Noir. The impacts are widely divergent, which show interesting patterns of genomic purging and the Hill-Robertson interference. Selfing reduces genomic heterozygosity, while cloning increases it, resulting in a "double U-shaped" site frequency spectrum (SFS). Crossing and cloning conceal while selfing purges most deleterious and structural burdens. Moreover, the close leakage of large-effect deleterious and structural variations in repulsion phases maintains heterozygous genomic regions in 4.3% of the grapevine genome after successive selfing for nine generations. Our study provides new insights into the genetic basis of clonal propagation and genomic breeding of clonal crops by purging deleterious variants while integrating beneficial variants through various reproductive systems.
Collapse
Affiliation(s)
- Hua Xiao
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yue Wang
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Wenwen Liu
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xiaoya Shi
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- College of Enology, Heyang Viti-Viniculture Station, Ningxia Helan Mountain's East Foothill Wine Experiment and Demonstration Station, Northwest A&F University, Yangling, China
| | - Siyang Huang
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Shuo Cao
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Qiming Long
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xu Wang
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Zhongjie Liu
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- Institute of Life and Health, China Resources Research Institute of Science and Technology, Hong Kong, China
| | - Xiaodong Xu
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yanling Peng
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | | | - Zhonghao Jiang
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Summaira Riaz
- San Joaquin Valley Agricultural Center, United States Department of Agriculture, Parlier, CA, USA
| | - Andrew M Walker
- San Joaquin Valley Agricultural Center, United States Department of Agriculture, Parlier, CA, USA
| | - Brandon S Gaut
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA, USA
| | - Sanwen Huang
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- National Key Laboratory of Tropical Crop Breeding, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Yongfeng Zhou
- National Key Laboratory of Tropical Crop Breeding, Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
- National Key Laboratory of Tropical Crop Breeding, Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China.
| |
Collapse
|
50
|
Guo Y, Charoenkwan P, Traisrisilp K, Piyamongkol W, Tongprasert F. Application of Digital Polymerase Chain Reaction (dPCR) in Non-Invasive Prenatal Testing (NIPT). Biomolecules 2025; 15:360. [PMID: 40149896 PMCID: PMC11940399 DOI: 10.3390/biom15030360] [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: 01/06/2025] [Revised: 02/17/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
This article reviews the current applications of the digital polymerase chain reaction (dPCR) in non-invasive prenatal testing (NIPT) and explores its potential to complement or surpass the capabilities of Next-Generation Sequencing (NGS) in prenatal testing. The growing incidence of genetic disorders in maternal-fetal medicine has intensified the demand for precise and accessible NIPT options, which aim to minimize the need for invasive prenatal diagnostic procedures. Cell-free fetal DNA (cffDNA), the core analyte in NIPT, is influenced by numerous factors such as maternal DNA contamination, placental health, and fragment degradation. dPCR, with its inherent precision and ability to detect low-abundance targets, demonstrates robustness against these interferences. Although NGS remains the gold standard due to its comprehensive diagnostic capabilities, its high costs limit widespread use, particularly in resource-limited settings. In contrast, dPCR provides comparable accuracy with lower complexity and expense, making it a promising alternative for prenatal testing.
Collapse
Affiliation(s)
- Ying Guo
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (Y.G.); (K.T.); (W.P.)
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Dali University, Dali 671000, China
| | - Pimlak Charoenkwan
- Department of Pediatrics, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand;
- Thalassemia and Hematology Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kuntharee Traisrisilp
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (Y.G.); (K.T.); (W.P.)
| | - Wirawit Piyamongkol
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (Y.G.); (K.T.); (W.P.)
| | - Fuanglada Tongprasert
- Department of Obstetrics and Gynaecology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (Y.G.); (K.T.); (W.P.)
| |
Collapse
|