1
|
Paudel D, Parrish SB, Peng Z, Parajuli S, Deng Z. A chromosome-scale and haplotype-resolved genome assembly of tetraploid blackberry ( Rubus L. subgenus Rubus Watson). HORTICULTURE RESEARCH 2025; 12:uhaf052. [PMID: 40271456 PMCID: PMC12015472 DOI: 10.1093/hr/uhaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 02/13/2025] [Indexed: 04/25/2025]
Abstract
Blackberries (Rubus spp.) are globally consumed and well known for their rich anthocyanin and antioxidant content and distinct flavors. Improving blackberries has been challenging due to genetic complexity of traits and limited genomic resources. The blackberry genome has been particularly challenging to assemble due to its polyploid nature. Here, we present the first chromosome-scale and haplotype-phased assembly for the primocane-fruiting, thornless tetraploid blackberry selection BL1 (Rubus L. subgenus Rubus Watson). The genome assembly was generated using Oxford Nanopore Technology and Hi-C scaffolding, resulting in a 919 Mb genome distributed across 27 pseudochromosomes, with an N50 of 35.73 Mb. This assembly covers >92% of the genome length and contains over 98% of complete BUSCOs. Approximately, 58% of the assembly consists of repetitive sequences, with long terminal repeats being the most abundant class. A total of 87,968 protein-coding genes were predicted, of which, 82% were functionally annotated. Genome mining and RNA-Seq analyses identified possible candidate genes and transcription factors related to thornlessness and the key structural genes and transcription factors for anthocyanin biosynthesis. Activator genes including PAP1 and TTG1 and repressor genes such as ANL2 and MYBPA1 play an important role in the fine tuning of anthocyanin production during blackberry development. Resequencing of seven tetraploid blackberry cultivars/selections with different horticultural characteristics revealed candidate genes that could impact fruiting habit and disease resistance/susceptibility. This tetraploid reference genome should provide a valuable resource for accelerating genetic analysis of blackberries and facilitating the development of new improved cultivars with enhanced horticultural and nutritional traits.
Collapse
Affiliation(s)
- Dev Paudel
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| | - S Brooks Parrish
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| | - Ze Peng
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| | - Saroj Parajuli
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| | - Zhanao Deng
- Gulf Coast Research and Education Center, Department of Environmental Horticulture, University of Florida, IFAS, 14625 County Road 672, Wimauma, FL 33598, USA
| |
Collapse
|
2
|
Sun H, Hao X, Liu H, Zhang S, Han J, Zhang Y, Liu T, Yang X, Wang H, Fan J, Guan Y, Peng N, Xie J, Xia H, Ji X, Xu Y, Zhang J, Wang J, Lan F, Zhang H, Gu X, He Y. Rare damaging variants in the sex differences of congenital heart disease: an exome sequencing study. EBioMedicine 2025; 116:105736. [PMID: 40347836 PMCID: PMC12139432 DOI: 10.1016/j.ebiom.2025.105736] [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: 08/19/2024] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Congenital heart disease (CHD) exhibits a marked male predominance in birth prevalence, yet the genetic mechanisms underlying this sex disparity remain poorly understood. This study investigates the contribution of rare damaging variants on autosomes and the X chromosome to sex differences in foetal CHD. METHODS Parents of foetuses with CHD were recruited for the study. Rare damaging variants were identified by analysing whole-exome sequencing data from foetus-parental trios, and their contributions to sex differences were estimated through case-control studies. Functional enrichment analysis was conducted to assess functional differences in genetic variants between sexes. FINDINGS 820 foetal probands with CHD were recruited, including 487 males and 333 females. We identified a significant enrichment of X-linked rare damaging variants, primarily driven by maternally inherited hemizygous variants (MIHVs) in male probands (OR = 1·84, P < 0·05), which accounted for 7·2% of male cases in our cohort. These variants were not found to be enriched in female probands. Additionally, X-linked rare damaging de novo variants (DNVs) were not enriched in either male or female probands (female probands: 1·8% versus female controls: 0·7%, P = 0·6789; no DNVs observed in males). Gene-level variant burden analysis revealed three X-linked CHD candidate genes: DCX, CACNA1F, and MAP3K15. Autosomal rare variants did not show significant differences in variant burdens between sexes. Notably, male probands showed specific functional enrichments in cilia-related pathways for autosomal recessive variants, as well as in chromatin remodelling and nervous system development pathways for autosomal DNVs. INTERPRETATION Male and female foetal CHD have significantly different genetic landscapes. The enrichment of X-linked rare damaging MIHVs in males provides a genetic explanation for the higher prevalence of CHD in males. This finding highlights the importance of incorporating sex-stratified approaches in clinical diagnostics and research. FUNDING Natural Science Foundation of China and Key research and development project of Hebei Province.
Collapse
Affiliation(s)
- Hairui Sun
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China
| | - Xiaoyan Hao
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Hankui Liu
- BGI-genomics, BGI-Shenzhen, Shenzhen, 518083, China
| | - Siyao Zhang
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Jiancheng Han
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Ye Zhang
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Tingting Liu
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Xian Yang
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Hairui Wang
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Jiaqi Fan
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Yuxuan Guan
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Ni Peng
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Jiaoyang Xie
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China
| | - Hongmei Xia
- Department of Ultrasound, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xueqin Ji
- Department of Ultrasound, Ningxia Women's and Children's Hospital, Beijing University First Hospital (Ningxia Hui Autonomous Region Maternal and Child Health Hospital), Yinchuan, 750004, China
| | - Yan Xu
- Rizhao People's Hospital, Shandong, 276827, China
| | | | - Jianbin Wang
- School of Life Sciences, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Feng Lan
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Hongjia Zhang
- Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, Beijing, 100069, China
| | - Xiaoyan Gu
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China; Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, Beijing, 100069, China.
| | - Yihua He
- Maternal-Fetal Medicine Center in Fetal Heart Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, 100029, China; Beijing Key Laboratory of Maternal-Fetal Medicine in Fetal Heart Disease, Beijing, 100029, China; Beijing Lab for Cardiovascular Precision Medicine, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
3
|
Kwan EM, Ng SWS, Tolmeijer SH, Emmett L, Sandhu S, Buteau JP, Iravani A, Joshua AM, Francis RJ, Subhash V, Lee ST, Scott AM, Martin AJ, Stockler MR, Donnellan G, Annala M, Herberts C, Davis ID, Hofman MS, Azad AA, Wyatt AW, TheraP Investigators and the ANZUP Cancer Trials Group. Lutetium-177-PSMA-617 or cabazitaxel in metastatic prostate cancer: circulating tumor DNA analysis of the randomized phase 2 TheraP trial. Nat Med 2025:10.1038/s41591-025-03704-9. [PMID: 40425844 DOI: 10.1038/s41591-025-03704-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 04/07/2025] [Indexed: 05/29/2025]
Abstract
The prostate-specific membrane antigen (PSMA)-targeted radioligand [¹⁷⁷Lu]Lu-PSMA-617 is a new standard treatment for metastatic castration-resistant prostate cancer (mCRPC), but predictive genomic biomarkers informing its rational use are unknown. We performed detailed dissection of prostate cancer driver genes across 290 serial plasma cell-free DNA samples from 180 molecular imaging-selected patients with mCRPC from the randomized TheraP trial of [¹⁷⁷Lu]Lu-PSMA-617 (n = 97) versus cabazitaxel chemotherapy (n = 83). The primary endpoint was PSA50 biochemical response, with secondary endpoints of progression-free survival (PFS) and overall survival (OS). In this post-hoc biomarker analysis, a low pretreatment circulating tumor DNA (ctDNA) fraction predicted a superior biochemical response (100% versus 58%, P = 0.0067) and PFS (median 14.7 versus 6.0 months; hazard ratio 0.12, P = 2.5 × 10-4) on [¹⁷⁷Lu]Lu-PSMA-617 independent of predictive PSMA-positron emission tomography imaging parameters, although this benefit did not extend to OS. Deleterious PTEN alterations were associated with worse PFS and OS on cabazitaxel, whereas ATM defects were observed in select patients with favorable [¹⁷⁷Lu]Lu-PSMA-617 outcomes. Comparing pretreatment and progression ctDNA revealed population flux but no evidence that alterations in individual mCRPC genes (or FOLH1) are dominant causes of acquired [¹⁷⁷Lu]Lu-PSMA-617 or cabazitaxel resistance. Our results nominate new candidate biomarkers for [¹⁷⁷Lu]Lu-PSMA-617 selection and ultimately expand the mCRPC predictive biomarker repertoire. We anticipate our ctDNA fraction-aware analytical framework will aid future precision management strategies for [¹⁷⁷Lu]Lu-PSMA-617 and other PSMA-targeted therapeutics. ClinicalTrials.gov identifier: NCT03392428 .
Collapse
Affiliation(s)
- Edmond M Kwan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia
- Biomedicine Discovery Institute Cancer Program, Prostate Cancer Research Group, Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria, Australia
| | - Sarah W S Ng
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sofie H Tolmeijer
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Shahneen Sandhu
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - James P Buteau
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Amir Iravani
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Division of Nuclear Medicine, Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anthony M Joshua
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Department of Medical Oncology, Kinghorn Cancer Centre, Sydney, New South Wales, Australia
| | - Roslyn J Francis
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Vinod Subhash
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group (ANZUP), Sydney, New South Wales, Australia
| | - Sze-Ting Lee
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Andrew M Scott
- Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Andrew J Martin
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, New South Wales, Australia
- Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| | - Martin R Stockler
- Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, New South Wales, Australia
| | - Gráinne Donnellan
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Matti Annala
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Cameron Herberts
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ian D Davis
- Eastern Health Clinical School, Monash University, Melbourne, Victoria, Australia.
- Cancer Services, Eastern Health, Melbourne, Victoria, Australia.
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Arun A Azad
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.
| | - Alexander W Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, Vancouver, British Columbia, Canada.
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada.
| | | |
Collapse
Collaborators
Tim Akhurst, Ramin Alipour, Dale L Bailey, Patricia Banks, Alexis Beaulieu, Louise Campbell, Wei Chua, Megan Crumbaker, Nattakorn Dhiantravan, Kate Ford, Craig Gedye, Jeffrey C Goh, Alex D Guminski, Anis Hamid, Mohammad B Haskali, Rodney J Hicks, Edward Hsiao, Terry Hung, Ian D Kirkwood, Grace Kong, Ailsa Langford, Nicola Lawrence, Jeremy Lewin, Peter Lin, Michael McCarthy, Margaret M McJannett, William McDonald, Kate Moodie, Declan G Murphy, Siobhan Ng, Andrew Nguyen, David A Pattison, David Pook, Izabella Pokorski, Shakher Ramdave, Nisha Rana, Aravind S Ravi Kumar, Andrew D Redfern, Paul Roach, Peter Roselt, Natalie K Rutherford, Javad Saghebi, Geoffrey Schembri, Lavinia Spain, Shalini Subramaniam, Thean Hsiang Tan, Sue Ping Thang, Paul Thomas, Ben Tran, John A Violet, Roslyn Wallace, Andrew Weickhardt, Scott G Williams, Sonia Yip, Alison Y Zhang,
Collapse
|
4
|
Moon SJ, Lee SH, Sim WH, Choi HS, Lee JS, Shim S. Haplotype-resolved chromosome-level genome sequence of Elsholtzia splendens (Nakai ex F.Maek.). Sci Data 2025; 12:827. [PMID: 40394069 PMCID: PMC12092835 DOI: 10.1038/s41597-025-05214-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 05/15/2025] [Indexed: 05/22/2025] Open
Abstract
Elsholtzia splendens, a perennial herb native to East Asia, is valued for its ornamental and medicinal uses, particularly in treating inflammatory and febrile conditions. Recent studies have highlighted its antibacterial, anti-inflammatory, antidepressant, antithrombotic, and lipid-lowering properties of its compounds. Additionally, E. splendens shows potential for phytoremediation owing to its ability to hyperaccumulate copper (Cu), lead (Pb), zinc (Zn), and cadmium (Cd). However, its role in remediation conflicts with its medicinal use because of the risk of heavy metal accumulation. Genome sequencing will be key to boosting beneficial compound production and reducing heavy metal risks. In this study, we generated a high-resolution, haplotype-resolved, chromosome-scale genome sequence of E. splendens using PacBio Revio long-read, Illumina short-read, and Hi-C sequencing technologies. The haplotype genome assemblies, spanned 275.4 and 265.0 Mbp with a scaffold N50 of 33.9 and 33.8 Mbp for haplotype 1 and 2, respectively. This assembly provides valuable insights into medicinal compound biosynthesis and supports genetic conservation efforts, facilitating future genetic and biotechnological applications of E. splendens for medicinal and ecological uses.
Collapse
Affiliation(s)
- Sung Jin Moon
- Department of Forest Resources, College of Forest and Environmental Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sae Hyun Lee
- Department of Agriculture, Forestry and Bioresources, College of Agriculture & Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Woo Hyun Sim
- Department of Forest Resources, College of Forest and Environmental Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Han Suk Choi
- Department of Forest Resources, College of Forest and Environmental Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ju Seok Lee
- Bio-evaluation Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju, 28116, Republic of Korea
| | - Sangrea Shim
- Department of Forest Resources, College of Forest and Environmental Sciences, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
5
|
Troester S, Eder T, Wukowits N, Piontek M, Fernández-Pernas P, Schmoellerl J, Haladik B, Manhart G, Allram M, Maurer-Granofszky M, Scheidegger N, Nebral K, Superti-Furga G, Meisel R, Bornhauser B, Valent P, Dworzak MN, Zuber J, Boztug K, Grebien F. Transcriptional and epigenetic rewiring by the NUP98::KDM5A fusion oncoprotein directly activates CDK12. Nat Commun 2025; 16:4656. [PMID: 40389480 PMCID: PMC12089343 DOI: 10.1038/s41467-025-59930-9] [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: 03/06/2024] [Accepted: 05/08/2025] [Indexed: 05/21/2025] Open
Abstract
Nucleoporin 98 (NUP98) fusion oncoproteins are strong drivers of pediatric acute myeloid leukemia (AML) with poor prognosis. Here we show that NUP98 fusion-expressing AML harbors an epigenetic signature that is characterized by increased accessibility of hematopoietic stem cell genes and enrichment of activating histone marks. We employ an AML model for ligand-induced degradation of the NUP98::KDM5A fusion oncoprotein to identify epigenetic programs and transcriptional targets that are directly regulated by NUP98::KDM5A through CUT&Tag and nascent RNA-seq. Orthogonal genome-wide CRISPR/Cas9 screening identifies 12 direct NUP98::KDM5A target genes, which are essential for AML cell growth. Among these, we validate cyclin-dependent kinase 12 (CDK12) as a druggable vulnerability in NUP98::KDM5A-expressing AML. In line with its role in the transcription of DNA damage repair genes, small-molecule-mediated CDK12 inactivation causes increased DNA damage, leading to AML cell death. Altogether, we show that NUP98::KDM5A directly regulates a core set of essential target genes and reveal CDK12 as an actionable vulnerability in AML with oncogenic NUP98 fusions.
Collapse
MESH Headings
- Humans
- Nuclear Pore Complex Proteins/metabolism
- Nuclear Pore Complex Proteins/genetics
- Oncogene Proteins, Fusion/metabolism
- Oncogene Proteins, Fusion/genetics
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Cyclin-Dependent Kinases/metabolism
- Cyclin-Dependent Kinases/genetics
- Epigenesis, Genetic
- Cell Line, Tumor
- Animals
- Transcription, Genetic
- Mice
- Gene Expression Regulation, Leukemic
- DNA Damage
- CRISPR-Cas Systems
- Retinoblastoma-Binding Protein 2
Collapse
Affiliation(s)
- Selina Troester
- Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Eder
- Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Nadja Wukowits
- Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Martin Piontek
- Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Pablo Fernández-Pernas
- Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Johannes Schmoellerl
- Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Ben Haladik
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Gabriele Manhart
- Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Melanie Allram
- Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | | | - Nastassja Scheidegger
- Division of Oncology and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Karin Nebral
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Labdia Labordiagnostik, Vienna, Austria
| | - Giulio Superti-Furga
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Beat Bornhauser
- Division of Oncology and Children's Research Centre, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseologay, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Michael N Dworzak
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
- Medical University of Vienna, Vienna BioCenter (VBC), Vienna, Austria
| | - Kaan Boztug
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Florian Grebien
- Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria.
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
6
|
Anton L, Cristancho AG, Ferguson B, Ravel J, Elovitz MA. Cervicovaginal microbiome alters transcriptomic and epigenomic signatures across cervicovaginal epithelial barriers. RESEARCH SQUARE 2025:rs.3.rs-6171614. [PMID: 40386438 PMCID: PMC12083688 DOI: 10.21203/rs.3.rs-6171614/v1] [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] [Indexed: 05/25/2025]
Abstract
Background The cervicovaginal microbiome plays a critical role in women's health, with microbial communities dominated by Lactobacillus species considered optimal. In contrast, the depletion of lactobacilli and the presence of a diverse array of strict and facultative anaerobes, such as Gardnerella vaginalis, have been linked with adverse reproductive outcomes. Despite these associations, the molecular mechanisms by which host-microbial interactions modulate cervical and vaginal epithelial function remains poorly understood. Results In this study, we used RNA sequencing to characterize the transcriptional response of cervicovaginal epithelial cells exposed to the culture supernatants of common vaginal bacteria. Our findings revealed that G. vaginalis culture supernatants upregulate genes associated with an activated innate immune response and increased cell death. Conversely, Lactobacillus crispatus culture supernatants induced transcriptional changes indicative of epigenomic modeling in ectocervical epithelial cells. Epigenomic modification by L. crispatus, was confirmed by ATAC-sequencing, which demonstrated reduced chromatin accessibility. Conclusions These results provide new insights into host-microbe interactions within the lower reproductive tract and suggests that modulating the vaginal microbiome could offer innovative therapeutic strategies to improve reproductive health.
Collapse
|
7
|
Urban JM, Gerbi SA, Spradling AC. Chromosome-scale scaffolds of the fungus gnat genome reveal multi-Mb-scale chromosome-folding interactions, centromeric enrichments of retrotransposons, and candidate telomere sequences. BMC Genomics 2025; 26:443. [PMID: 40325439 PMCID: PMC12051294 DOI: 10.1186/s12864-025-11573-2] [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: 01/09/2025] [Accepted: 04/04/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND The lower Dipteran fungus gnat, Bradysia (aka Sciara) coprophila, has compelling chromosome biology. Paternal chromosomes are eliminated during male meiosis I and both maternal X sister chromatids are retained in male meiosis II. Embryos start with three copies of the X chromosome, but 1-2 copies are eliminated from somatic cells as part of sex determination, and one is eliminated in the germline to restore diploidy. In addition, there is gene amplification in larval polytene chromosomes, and the X polytene chromosome folds back on itself mediated by extremely long-range interactions between three loci. These developmentally normal events present opportunities to study chromosome behaviors that are unusual in other systems. Moreover, little is known about the centromeric and telomeric sequences of lower Dipterans in general, and there are recent claims of horizontally-transferred genes in fungus gnats. Overall, there is a pressing need to learn more about the fungus gnat chromosome sequences. RESULTS We produced the first chromosome-scale models of the X and autosomal chromosomes where each somatic chromosome is represented by a single scaffold. Extensive analysis supports the chromosome identity and structural accuracy of the scaffolds, demonstrating they are co-linear with historical polytene maps, consistent with evolutionary expectations, and have accurate centromere positions, chromosome lengths, and copy numbers. The positions of alleged horizontally-transferred genes in the nuclear chromosomes were broadly confirmed by genomic analyses of the chromosome scaffolds using Hi-C and single-molecule long-read datasets. The chromosomal context of repeats shows family-specific biases, such as retrotransposons correlated with the centromeres. Moreover, scaffold termini were enriched with arrays of retrotransposon-related sequence as well as nucleosome-length (~ 175 bp) satellite repeats. Finally, the Hi-C data captured Mb-scale physical interactions on the X chromosome that are seen in polytene spreads, and we characterize these interesting "fold-back regions" at the sequence level for the first time. CONCLUSIONS The chromosome scaffolds were shown to be of exceptional quality, including loci harboring horizontally-transferred genes. Repeat analyses demonstrate family-specific biases and telomere repeat candidates. Hi-C analyses revealed the sequences of ultra-long-range interactions on the X chromosome. The chromosome-scale scaffolds pave the way for further studies of the unusual chromosome movements in Bradysia coprophila.
Collapse
Affiliation(s)
- John M Urban
- Carnegie Institution for Science, Department of Embryology, Howard Hughes Medical Institute Research Laboratories, 3520 San Martin Drive, Baltimore, MD, 21218, USA.
| | - Susan A Gerbi
- Division of Biology and Medicine, Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02912, USA
| | - Allan C Spradling
- Carnegie Institution for Science, Department of Embryology, Howard Hughes Medical Institute Research Laboratories, 3520 San Martin Drive, Baltimore, MD, 21218, USA
| |
Collapse
|
8
|
Verdu Schlie A, Leitch A, Arismendi MI, Stok C, Castro Leal A, Parry DA, Marcondes Lerario A, Harley ME, Lucheze B, Carroll PL, Musialik KI, Auer JMT, Martin CA, Gerasimavicius L, Quigley AJ, de Menezes Correia-Deur JE, Marsh JA, Reijns MAM, Lampe AK, Jackson AP, Jorge AAL, Tamayo-Orrego L. CDK4 loss-of-function mutations cause microcephaly and short stature. Genes Dev 2025; 39:634-651. [PMID: 40210435 PMCID: PMC7617628 DOI: 10.1101/gad.352311.124] [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: 09/20/2024] [Accepted: 03/06/2025] [Indexed: 04/12/2025]
Abstract
Cell number is a major determinant of organism size in mammals. In humans, gene mutations in cell cycle components result in restricted growth through reduced cell numbers. Here we identified biallelic mutations in CDK4 as a cause of microcephaly and short stature. CDK4 encodes a key cell cycle kinase that associates with D-type cyclins during G1 of the cell cycle to promote S-phase entry and cell proliferation through retinoblastoma (RB) phosphorylation. CDK4 and CDK6 are believed to be functionally redundant and are targeted jointly by chemotherapeutic CDK4/6 inhibitors. Using molecular and cell biology approaches, we show that functional CDK4 protein is not detectable in cells with CDK4 mutations. Cells display impaired RB phosphorylation in G1, leading to G1/S-phase transition defects and reduced cell proliferation, consistent with complete loss of cellular CDK4 enzymatic activity. Together, these findings demonstrate that CDK4 is itself required for cell proliferation, human growth, and brain size determination during development.
Collapse
Affiliation(s)
- Aitana Verdu Schlie
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Andrea Leitch
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Maria Izabel Arismendi
- Genetic Endocrinology Unit (LIM25), Endocrinology Division, Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), São Paulo 01246-903, Brazil
| | - Colin Stok
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Andrea Castro Leal
- Department of Integrated Health, State University of Para, Santarem 68010-200, Brazil
| | - David A Parry
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Antonio Marcondes Lerario
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Margaret E Harley
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Bruna Lucheze
- Genetic Endocrinology Unit (LIM25), Endocrinology Division, Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), São Paulo 01246-903, Brazil
| | - Paula L Carroll
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Kamila I Musialik
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Julia M T Auer
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Carol-Anne Martin
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Lukas Gerasimavicius
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Alan J Quigley
- Paediatric Imaging Department, Royal Hospital for Children and Young People, Edinburgh EH16 4TJ, United Kingdom
| | - Joya Emilie de Menezes Correia-Deur
- Genetic Endocrinology Unit (LIM25), Endocrinology Division, Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), São Paulo 01246-903, Brazil
| | - Joseph A Marsh
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Martin A M Reijns
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Anne K Lampe
- South East of Scotland Clinical Genetics Service, Western General Hospital, Edinburgh EH4 2XU, United Kingdom
| | - Andrew P Jackson
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom;
| | - Alexander A L Jorge
- Genetic Endocrinology Unit (LIM25), Endocrinology Division, Faculdade de Medicina da Universidade de São Paulo (HC-FMUSP), São Paulo 01246-903, Brazil;
| | - Lukas Tamayo-Orrego
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| |
Collapse
|
9
|
Kumar S, Biswas S, Agarwal S, Sheikh S, Ashraf A, Swaroop S, Mehta S, Vasant S, Pradhan D, Nayak B, Shalimar. Next-Generation Sequencing Identifies Novel Germline Mutations in Patients with Budd-Chiari Syndrome-Associated Hepatocellular Carcinoma. Dig Dis Sci 2025; 70:1788-1798. [PMID: 40021601 DOI: 10.1007/s10620-025-08942-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
INTRODUCTION Budd-Chiari syndrome-hepatocellular carcinoma (BCS-HCC) is uncommon and its molecular pathogenesis is poorly understood. In this study, we aimed to investigate the genomic landscape of BCS-HCC through whole exome sequencing (WES) to elucidate the cellular and molecular pathways involved in its pathogenesis. METHODOLOGY We enrolled BCS-HCC (n = 13) and BCS alone (n = 73) patients. WES was performed using the Twist Comprehensive Exome kit on the Illumina platform, followed by quality checks and analysis using the GATK pipeline. Pathogenic/likely pathogenic variants were filtered out from the exonic part of the annotated variant calling file. rsIDs and Cosmic IDs (catalogue of somatic mutations in cancer) were assigned using dbSNP and Cosmic ID databases. Kyoto Encyclopaedia of Genes and Genomes (KEGG) and Gene ontology analysis were done for pathogenic gene variants. RESULTS We observed 1849 significant mutations in 305 genes in BCS-HCC patients, including missense, Indel, and frameshift mutations. Missense variants were more common than frameshift and indels in all subjects. The pathogenic mutations were found in 34 genes-cancer-causing (18 genes) and disease-causing (16 genes, both BCS or BCS-HCC) as per COSMIC cancer gene census. Pathogenic mutations were frequently observed in the mucin family genes including MUC3A, MUC4, MUC6, and MUC16 in BCS-HCC subjects. Changes in extracellular matrix and glycosylation were observed in gene ontology analysis of the genes having pathogenic variants. CONCLUSION Mutations in the mucin gene family including other cancer-causing genes were associated with BCS-HCC in our cohort. Larger, multicentric studies with regional and ethnic diversity are required to validate these findings.
Collapse
Affiliation(s)
- Sonu Kumar
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Sagnik Biswas
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Samagra Agarwal
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Sabreena Sheikh
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Anzar Ashraf
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Shekhar Swaroop
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Shubham Mehta
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Shrinidhi Vasant
- Department of Biotechnology, Banasthali Vidyapith, Tonk, Rajasthan, India
| | - Dibyabhabha Pradhan
- Central Core Research Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Baibaswata Nayak
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India
| | - Shalimar
- Department of Gastroenterology and Human Nutrition Unit, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
10
|
Brunharo CA, Short AW, Bobadilla LK, Streisfeld MA. The Genome of Lolium multiflorum Reveals the Genetic Architecture of Paraquat Resistance. Mol Ecol 2025; 34:e17775. [PMID: 40285737 PMCID: PMC12051776 DOI: 10.1111/mec.17775] [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: 09/15/2024] [Revised: 04/03/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
Herbicide resistance in agricultural weeds has become one of the greatest challenges for sustainable crop production. The repeated evolution of herbicide resistance provides an excellent opportunity to study the genetic and physiological basis of the resistance phenotype and the evolutionary responses to human-mediated selection pressures. Lolium multiflorum is a ubiquitous weed that has evolved herbicide resistance repeatedly around the world in various cropping systems. We assembled and annotated a chromosome-scale genome for L. multiflorum and elucidated the genetic architecture of paraquat resistance by performing quantitative trait locus analysis, genome-wide association studies, genetic divergence analysis and transcriptome analyses from paraquat-resistant and -susceptible L. multiflorum plants. We identified two regions on chromosome 5 that were associated with paraquat resistance. These regions both showed evidence for positive selection among the resistant populations we sampled, but the effects of this selection on the genome differed, implying a complex evolutionary history. In addition, these regions contained candidate genes that encoded cellular transport functions, including a novel multidrug and toxin extrusion (MATE) protein and a cation transporter previously shown to interact with polyamines. Given that L. multiflorum is a weed and a cultivated crop species, the genomic resources generated will prove valuable to a wide spectrum of the plant science community. Our work contributes to a growing body of knowledge on the underlying evolutionary and ecological dynamics of rapid adaptation to strong anthropogenic selection pressure that could help initiate efforts to improve weed management practices in the long term for a more sustainable agriculture.
Collapse
Affiliation(s)
- Caio A. Brunharo
- Department of Plant ScienceThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| | - Aidan W. Short
- Institute of Ecology and Evolution, University of OregonEugeneOregonUSA
| | | | | |
Collapse
|
11
|
Guseva M, Shirokova N, Kapitonova O, Gnetetskaya V, Blagodatskikh K, Tarasova J, Kaimonov V, Korbut A, Musatova E. Application of Whole-Exome Sequencing (WES) for Prenatal Determination of Causes of Fetal Abnormalities. Genes (Basel) 2025; 16:547. [PMID: 40428369 PMCID: PMC12111140 DOI: 10.3390/genes16050547] [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: 03/25/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Fetal abnormalities are major issues in prenatal medicine. They affect the predicted pregnancy outcome and entail a risk of future recurrent adverse events in a particular couple. In order to clarify the possible outcomes of a specific pregnancy and subsequent ones, it is of the utmost importance to determine the causes of observed fetal abnormalities. Routine laboratory techniques sometimes fail to identify their cause because they are mainly intended for the detection of chromosomal disorders. Over recent years, single-gene disorders have increasingly been regarded as probable causes of fetal abnormalities, and next-generation sequencing (NGS) technologies have been adopted to detect them. This article provides the findings of applying whole-exome sequencing (WES) for prenatal diagnosis. It is aimed at identifying the causes of various fetus abnormalities with a normal molecular karyotype. The diagnostic value of this technique is shown based on the completeness of clinical patterns, abnormality types, and the ability to simultaneously examine the fetus and the parents. Consequently, WES revealed causative variants in 27.27% of cases, which encourages the consideration of applying this technique as part of a state-of-the-art multiple congenital malformation prenatal diagnosis algorithm.
Collapse
Affiliation(s)
- Margarita Guseva
- The Center of Genetics and Reproductive Medicine “Genetico” JSC, 119571 Moscow, Russia; (N.S.); (O.K.); (K.B.); (V.K.); (A.K.); (E.M.)
| | - Natalya Shirokova
- The Center of Genetics and Reproductive Medicine “Genetico” JSC, 119571 Moscow, Russia; (N.S.); (O.K.); (K.B.); (V.K.); (A.K.); (E.M.)
| | - Olga Kapitonova
- The Center of Genetics and Reproductive Medicine “Genetico” JSC, 119571 Moscow, Russia; (N.S.); (O.K.); (K.B.); (V.K.); (A.K.); (E.M.)
| | | | - Konstantin Blagodatskikh
- The Center of Genetics and Reproductive Medicine “Genetico” JSC, 119571 Moscow, Russia; (N.S.); (O.K.); (K.B.); (V.K.); (A.K.); (E.M.)
| | - Julia Tarasova
- Clinic “Mother and Child” Savelovskaya, 127015 Moscow, Russia
| | - Vladimir Kaimonov
- The Center of Genetics and Reproductive Medicine “Genetico” JSC, 119571 Moscow, Russia; (N.S.); (O.K.); (K.B.); (V.K.); (A.K.); (E.M.)
| | - Alina Korbut
- The Center of Genetics and Reproductive Medicine “Genetico” JSC, 119571 Moscow, Russia; (N.S.); (O.K.); (K.B.); (V.K.); (A.K.); (E.M.)
| | - Elizaveta Musatova
- The Center of Genetics and Reproductive Medicine “Genetico” JSC, 119571 Moscow, Russia; (N.S.); (O.K.); (K.B.); (V.K.); (A.K.); (E.M.)
- Federal State Budgetary Educational Institution, Further Professional Education “Russian Medical Academy of Continuous Professional Education”, the Ministry of Healthcare of the Russian Federation, 125993 Moscow, Russia
| |
Collapse
|
12
|
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
|
13
|
Allen-Brady K, Moore B, Verrilli LE, Alvord MA, Kern M, Camp N, Kelley K, Letourneau J, Cannon-Albright L, Yandell M, Johnstone EB, Welt CK. Breast Cancer Is Increased in Women With Primary Ovarian Insufficiency. J Clin Endocrinol Metab 2025; 110:e1678-e1686. [PMID: 38996041 PMCID: PMC12012772 DOI: 10.1210/clinem/dgae480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/26/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
CONTEXT DNA damage/repair gene variants are associated with both primary ovarian insufficiency (POI) and cancer risk. OBJECTIVE We hypothesized that a subset of women with POI and family members would have increased risk for cancer. DESIGN Case-control population-based study using records from 1995 to 2022. SETTING Two major Utah academic health care systems serving 85% of the state. SUBJECTS Women with POI (n = 613) were identified using International Classification of Diseases codes and reviewed for accuracy. Relatives were linked using the Utah Population Database. INTERVENTION Cancer diagnoses were identified using the Utah Cancer Registry. MAIN OUTCOME MEASURES The relative risk of cancer in women with POI and relatives was estimated by comparison to population rates. Whole genome sequencing was performed on a subset of women. RESULTS Breast cancer was increased in women with POI (OR, 2.20; 95% CI, 1.30-3.47; P = .0023) and there was a nominally significant increase in ovarian cancer. Probands with POI were 36.5 ± 4.3 years and 59.5 ± 12.7 years when diagnosed with POI and cancer, respectively. Causal and candidate gene variants for cancer and POI were identified. Among second-degree relatives of these women, there was an increased risk of breast (OR, 1.28; 95% CI, 1.08-1.52; P = .0078) and colon cancer (OR, 1.50; 95% CI, 1.14-1.94; P = .0036). Prostate cancer was increased in first- (OR, 1.64; 95% CI, 1.18-2.23; P = .0026), second- (OR, 1.54; 95% CI, 1.32-1.79; P < .001), and third-degree relatives (OR, 1.33; 95% CI, 1.20-1.48; P < .001). CONCLUSION Data suggest common genetic risk for POI and reproductive cancers. Tools are needed to predict cancer risk in women with POI and potentially to counsel about risks of hormone replacement therapy.
Collapse
Affiliation(s)
- Kristina Allen-Brady
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Barry Moore
- Utah Center for Genetic Discovery, Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Lauren E Verrilli
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Intermountain Healthcare, Murray, UT 84107, USA
| | - Margaret A Alvord
- Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Marina Kern
- Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Nicola Camp
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Kristen Kelley
- Huntsman Cancer Institute and Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | - Joseph Letourneau
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Lisa Cannon-Albright
- Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | - Mark Yandell
- Utah Center for Genetic Discovery, Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Erica B Johnstone
- Division of Reproductive Endocrinology and Infertility, Department of Obstetrics and Gynecology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Corrine K Welt
- Division of Endocrinology, Metabolism and Diabetes, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
14
|
Rochus CM, Steensma MJ, Bink MCAM, Huisman AE, Harlizius B, Derks MFL, Crooijmans RPMA, Ducro BJ, Bijma P, Groenen MAM, Mulder HA. Estimating mutation rate and characterising single nucleotide de novo mutations in pigs. Genet Sel Evol 2025; 57:21. [PMID: 40229661 PMCID: PMC11995543 DOI: 10.1186/s12711-025-00967-1] [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: 06/03/2024] [Accepted: 04/03/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Direct estimates of mutation rates in humans have changed our understanding of evolutionary timing and de novo mutations (DNM) have been associated with several developmental disorders in humans. Livestock species, including pigs, can contribute to the study of DNM because of their ideal population structure and routine phenotype collection. In principle, there is the potential for livestock populations to quickly accumulate new genetic variants because of short generation intervals and high selection intensity. However, the impact of DNM on the fitness of individuals is not known and with current genomic selection programs they cannot contribute to estimated breeding values. The aims of our project were to detect and validate single nucleotide DNM in two commercial pig breeding lines, estimate the single nucleotide mutation rate, and characterise DNM. RESULTS We sequenced (150 bp paired end reads, 30X coverage) 46 pig trios from two commercial lines. Single nucleotide DNM were detected using a trio-aware method. We defined candidate DNM as single nucleotide variants (SNVs) found in heterozygous state in trio-offspring with both trio-parents homozygous for the reference allele. In this study, we estimate a lower threshold of the DNM rate in pigs of 6.3 × 10-9 per site per gamete. Our findings are consistent with those from other mammals and those published for a small number of livestock species. Most DNM we detected were in introns (47%) and intergenic regions (49%). The mutational spectrum in pigs differs from that in humans and we found several DNM predicted to have an effect on animal's fitness based on the base pair change and their location in the genome. CONCLUSIONS With this study, we have generated fundamental knowledge on mutation rate in a non-primate species and identified DNM that could have an impact on the fitness of individuals.
Collapse
Affiliation(s)
- Christina M Rochus
- Wageningen University & Research Animal Breeding and Genomics, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
- The University of Edinburgh, The Roslin Institute Easter Bush Campus, Midlothian, EH25 9RG, Scotland
| | - Marije J Steensma
- Wageningen University & Research Animal Breeding and Genomics, P.O. Box 338, 6700 AH, Wageningen, The Netherlands.
| | - Marco C A M Bink
- Hendrix Genetics, P.O. Box 114, 5830 AC, Boxmeer, The Netherlands
| | - Abe E Huisman
- Hendrix Genetics, P.O. Box 114, 5830 AC, Boxmeer, The Netherlands
| | - Barbara Harlizius
- Topigs Norsvin Research Center, Meerendonkweg 25, 5216 TZ, Den Bosch, The Netherlands
| | - Martijn F L Derks
- Wageningen University & Research Animal Breeding and Genomics, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
- Topigs Norsvin Research Center, Meerendonkweg 25, 5216 TZ, Den Bosch, The Netherlands
| | - Richard P M A Crooijmans
- Wageningen University & Research Animal Breeding and Genomics, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| | - Bart J Ducro
- Wageningen University & Research Animal Breeding and Genomics, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| | - Piter Bijma
- Wageningen University & Research Animal Breeding and Genomics, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| | - Martien A M Groenen
- Wageningen University & Research Animal Breeding and Genomics, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| | - Han A Mulder
- Wageningen University & Research Animal Breeding and Genomics, P.O. Box 338, 6700 AH, Wageningen, The Netherlands
| |
Collapse
|
15
|
Black GS, Huang X, Qiao Y, Moos P, Sampath D, Stephens DM, Woyach JA, Marth GT. Long-read single-cell RNA sequencing enables the study of cancer subclone-specific genotypes and phenotypes in chronic lymphocytic leukemia. Genome Res 2025; 35:686-697. [PMID: 39965935 PMCID: PMC12047255 DOI: 10.1101/gr.279049.124] [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: 03/15/2024] [Accepted: 01/30/2025] [Indexed: 02/20/2025]
Abstract
Bruton tyrosine kinase (BTK) inhibitors are effective for the treatment of chronic lymphocytic leukemia (CLL) due to BTK's role in B cell survival and proliferation. Treatment resistance is most commonly caused by the emergence of the hallmark BTK C481S mutation that inhibits drug binding. In this study, we aimed to investigate cancer subclones harboring a BTK C481S mutation and identify cells with co-occurring CLL driver mutations. In addition, we sought to determine whether BTK-mutated subclones exhibit distinct transcriptomic behavior when compared to other cancer subclones. To achieve these goals, we use scBayes, which integrates bulk DNA sequencing and single-cell RNA sequencing (scRNA-seq) data to genotype individual cells for subclone-defining mutations. Although the most common approach for scRNA-seq includes short-read sequencing, transcript coverage is limited due to the vast majority of the reads being concentrated at the priming end of the transcript. Here, we utilized MAS-seq, a long-read scRNA-seq technology, to substantially increase transcript coverage and expand the set of informative mutations to link cells to cancer subclones in six CLL patients who acquired BTK C481S mutations during BTK inhibitor treatment. In two patients who developed two independent BTK-mutated subclones, we find that most BTK-mutated cells have an additional CLL driver gene mutation. When examining subclone-specific gene expression, we find that in one patient, BTK-mutated subclones are transcriptionally distinct from the rest of the malignant B cell population with an overexpression of CLL-relevant genes.
Collapse
Affiliation(s)
- Gage S Black
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA
| | - Xiaomeng Huang
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA
| | - Yi Qiao
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA
| | - Philip Moos
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, Utah 84112, USA
| | - Deepa Sampath
- Department of Hematopoietic Biology and Malignancy, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Deborah M Stephens
- Division of Hematology, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | - Jennifer A Woyach
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA
| | - Gabor T Marth
- Department of Human Genetics, University of Utah, Salt Lake City, Utah 84112, USA;
| |
Collapse
|
16
|
Wang Y, Yu A, Gao Z, Yuan X, Du X, Shi P, Guan H, Wen S, Wang H, Wang L, Fan B, Liu Z. TET2 gene mutation status associated with poor prognosis of transition zone prostate cancer: a retrospective cohort study based on whole exome sequencing and machine learning models. Front Endocrinol (Lausanne) 2025; 16:1568665. [PMID: 40297177 PMCID: PMC12034557 DOI: 10.3389/fendo.2025.1568665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Prostate cancer (PCa) in the transition zone (TZ) is uncommon and often poses challenges for early diagnosis, but its genomic determinants and therapeutic vulnerabilities remain poorly characterized. Methods Tumor mutational landscape was characterized in nine patients with TZ PCa, identifying somatic variants through whole-exome sequencing (WES). Novel candidate variants relevant to driver gene were selected using rare-variant burden analysis. Kaplan-Meier curves with log-rank testing and Cox regression models were applied to evaluate the prognostic significance of selected mutant driver gene and clinicopathological characteristics in a cohort of 132 patients with TZ PCa. Significant prognostic determinants were integrated into a validated nomogram for individualized prediction of 3-, 4-, and 5-year biochemical recurrence-free survival (BRFS) and overall survival (OS) probabilities. Eight machine learning algorithms were employed to develop BRFS and OS prediction models in a cohort. Results A total of 5,036 somatic single nucleotide variants (SNVs) and 587 somatic insertion and deletion (INDELs) were discovered. Among eight driver gene mutations which were verified through Sanger sequencing, TET2 gene, with high mutation frequency and potential targeted drug relevance (bromodomain inhibitors and DOT1L inhibitors) was selected for further validation. Retrospective cohort study demonstrated that TET2 mutant status was significantly associated with Gleason score (p = 0.004) and distant metastasis (p = 0.002). Furthermore, TET2 mutant status was significantly correlated with inferior BRFS and OS and served as an independent predictor. Comparative evaluation of eight algorithms revealed the GBM model achieved superior discriminative ability for BRFS (AUC for 3-year: 0.752, 4-year: 0.786, 5-year: 0.796). The predictive model based on the GBM machine learning algorithm achieved the best predictive performance for OS (AUC for 3-year: 0.838, 4-year: 0.915, 5-year: 0.868). The constructed predictive nomogram provided evidence that TET2 mutant status integration conferred statistically significant improvements in model accuracy and clinical predictive value. Conclusion Our study elucidated the distinct genetic basis of prostate cancer in the transition zone and identified TET2 mutation as an independent prognostic determinant in TZ PCa. However, the limited sample size of this study necessitates cautious interpretation of these findings, and further validation in larger cohorts is warranted to confirm their generalizability.
Collapse
Affiliation(s)
- Yutong Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Colorectal Surgery, Dalian Municipal Central Hospital, Dalian, Liaoning, China
| | - Ailing Yu
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- First Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Ziping Gao
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiaoying Yuan
- Department of Anatomy, College of Basic Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Xiaochen Du
- College of Humanities and Social Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Peng Shi
- Second Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Haoyun Guan
- Second Clinical College, Dalian Medical University, Dalian, Liaoning, China
| | - Shuang Wen
- Department of Pathology, Dalian Friendship Hospital, Dalian, China
| | - Honglong Wang
- Department of Pathology, Dalian Friendship Hospital, Dalian, China
| | - Liang Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bo Fan
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhiyu Liu
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Department of Urology, Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
17
|
Beare PA, Lack J, Anderson M, Cockrell DC, Heinzen RA, Long CM. Whole genome sequence of Coxiella burnetii Nine Mile II, clone 4, ∆ cbu0533 mutant strain, an alternative for safer laboratory use. Microbiol Resour Announc 2025; 14:e0073024. [PMID: 40062763 PMCID: PMC11984202 DOI: 10.1128/mra.00730-24] [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: 07/17/2024] [Accepted: 01/23/2025] [Indexed: 04/11/2025] Open
Abstract
Here, we determined the genome sequence of the Coxiella burnetii Nine Mile II (NMII), RSA439, clone 4, ∆cbu0533 mutant strain. This strain was engineered as a potential laboratory replacement for the wild-type C. burnetii NMII, which can undergo lipopolysaccharide elongation in specific growth conditions due to cbu0533 mutation reversion.
Collapse
Affiliation(s)
- Paul A. Beare
- Genomics Section, Research Technologies Branch, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, Montana, USA
| | - Justin Lack
- Integrated Data Sciences Section, Research Technologies Branch, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew Anderson
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, Montana, USA
| | - Diane C. Cockrell
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, Montana, USA
| | - Robert A. Heinzen
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, Montana, USA
| | - Carrie M. Long
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Hamilton, Montana, USA
| |
Collapse
|
18
|
Chen D, Lei T, Wang Y, Yu Z, Liu S, Ye L, Li W, Yang Q, Jin H, Liu F, Li Y. Isolation and Characterization of the Adamantinomatous Craniopharyngioma Primary Cells with Cancer-Associated Fibroblast Features. Biomedicines 2025; 13:912. [PMID: 40299526 PMCID: PMC12025058 DOI: 10.3390/biomedicines13040912] [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: 03/03/2025] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
Backgrounds: Adamantinomatous craniopharyngiomas (ACPs) are benign intracranial tumors that behave aggressively due to their location, infiltration of the surrounding nervous tissue and high capacity for recurrence. In this study, we aimed to construct ACP primary cell models for further investigation of tumorigenic and recurrent mechanisms. Methods: Primary cells were isolated from primary (one case) and recurrent (one case) ACP. Short tandem repeat (STR) analysis was used to clarify the identity of the ACP primary cells we isolated. Whole exome sequencing (WES), immunofluorescence (IF) and immunohistochemistry (IHC) were performed on primary cells and corresponding ACP tissues, to determine the mutational profile and to clarify the tissue origin and phenotypic of primary cells. Transcriptome RNA-seq was performed to obtain the gene expression characteristics of ACP primary cells. Subsequently, a heterotopic ACP xenograft mouse model was established to confirm the tumorigenesis capacity of ACP primary cells. Results: ACP primary cells were successfully cultured. The genetic variants were similar to the original tumor tissue, and they owned expression of cancer-associated fibroblast (CAF) markers (FSP1/S100A4, Vimentin) and nuclear translocation β-catenin. Meanwhile, they had an high level expression of extracellular matrix components (Fibronectin). The tumor formation ability of ACP primary cells was verified. The transcriptional signatures of ACP primary cells were also explored. Conclusions: We successfully isolated and characterized ACP primary cells that acquired multiple CAF features and demonstrated stable propagation through dozens of passages. These PDC models laid the foundation for further research on ACP.
Collapse
Affiliation(s)
- Dongting Chen
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| | - Ting Lei
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China; (T.L.)
| | - Yulin Wang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| | - Zaitao Yu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China; (T.L.)
| | - Siqi Liu
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| | - Ling Ye
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| | - Wanfang Li
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Union-Genius Pharmaceutical Technology Development Co., Ltd., Beijing 100176, China
| | - Qin Yang
- Beijing Union-Genius Pharmaceutical Technology Development Co., Ltd., Beijing 100176, China
| | - Hongtao Jin
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
- Beijing Union-Genius Pharmaceutical Technology Development Co., Ltd., Beijing 100176, China
| | - Fangjun Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China; (T.L.)
| | - Yan Li
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (D.C.)
| |
Collapse
|
19
|
Armstrong EE, Carey SB, Harkess A, Zenato Lazzari G, Solari KA, Maldonado JE, Fleischer RC, Aziz N, Walsh P, Koepfli KP, Eizirik E, Petrov DA, Campana MG. Parameterizing Pantherinae: De Novo Mutation Rate Estimates from Panthera and Neofelis Pedigrees. Genome Biol Evol 2025; 17:evaf060. [PMID: 40171701 PMCID: PMC11997302 DOI: 10.1093/gbe/evaf060] [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: 05/28/2024] [Revised: 03/12/2025] [Accepted: 03/23/2025] [Indexed: 04/04/2025] Open
Abstract
Estimates of de novo mutation rates are essential for phylogenetic and demographic analyses, but their inference has previously been impeded by high error rates in sequence data and uncertainty in the fossil record. Here, we directly estimate de novo germline mutation rates for all extant members of Panthera, as well as the closely related outgroup Neofelis nebulosa, using pedigrees. We use a previously validated pipeline (RatesTools) to calculate mutation rates for each species and subsequently explore the impacts of the novel rates on historic effective population size estimates in each of these charismatic felids of conservation concern. Importantly, we find that the choice of reference genome, the data type and coverage, and the individual all impact estimates of the mutation rate, but these can be largely ameliorated through extensive manual curation. Despite these stochastic effects, manual validation of de novo mutation candidates permitted the reliable inference of pantherine mutation rates. We inferred that base pair mutation rates for all species fell between 3.6 × 10-9 and 7.6 × 10-9 per generation per base pair (mean 5.5 × 10-9 ± 1.7 × 10-9 across Pantherinae at a mean parental age of 5.5 years). Similar to other studies, we show a positive trend of mean parental age with mutation rate and our inferred rates are well within the expected range for other mammals.
Collapse
Affiliation(s)
- Ellie E Armstrong
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Biology, Washington State University, Pullman, WA, USA
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, Riverside, CA, USA
- Center for Conservation Genomics, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| | - Sarah B Carey
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Alex Harkess
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Gabriele Zenato Lazzari
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | - Jesús E Maldonado
- Center for Conservation Genomics, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| | - Robert C Fleischer
- Center for Conservation Genomics, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| | - Neel Aziz
- Department of Animal Care Sciences, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| | - Patricia Walsh
- Department of Animal Care Sciences, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| | - Klaus-Peter Koepfli
- Smithsonian-Mason School of Conservation, George Mason University, Front Royal, VA, USA
- Center for Species Survival, Smithsonian's National Zoo and Conservation Biology Institute, Front Royal, VA, USA
| | - Eduardo Eizirik
- School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Dmitri A Petrov
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Michael G Campana
- Center for Conservation Genomics, Smithsonian's National Zoo and Conservation Biology Institute, Washington, DC, USA
| |
Collapse
|
20
|
Lee S, Roh J, Park JS, Tuncay IO, Lee W, Kim JA, Oh BBL, Shin JY, Lee JS, Ju YS, Kim R, Park S, Koo J, Park H, Lim J, Connolly-Strong E, Kim TH, Choi YW, Ahn MS, Lee HW, Kim S, Kim JH, Kwon M. Target-Enhanced Whole-Genome Sequencing Shows Clinical Validity Equivalent to Commercially Available Targeted Oncology Panel. Cancer Res Treat 2025; 57:350-361. [PMID: 39300929 PMCID: PMC12016826 DOI: 10.4143/crt.2024.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
PURPOSE Cancer poses a significant global health challenge, demanding precise genomic testing for individualized treatment strategies. Targeted-panel sequencing (TPS) has improved personalized oncology but often lacks comprehensive coverage of crucial cancer alterations. Whole-genome sequencing (WGS) addresses this gap, offering extensive genomic testing. This study demonstrates the medical potential of WGS. MATERIALS AND METHODS This study evaluates target-enhanced WGS (TE-WGS), a clinical-grade WGS method sequencing both cancer and matched normal tissues. Forty-nine patients with various solid cancer types underwent both TE-WGS and TruSight Oncology 500 (TSO500), one of the mainstream TPS approaches. RESULTS TE-WGS detected all variants reported by TSO500 (100%, 498/498). A high correlation in variant allele fractions was observed between TE-WGS and TSO500 (r=0.978). Notably, 223 variants (44.8%) within the common set were discerned exclusively by TE-WGS in peripheral blood, suggesting their germline origin. Conversely, the remaining subset of 275 variants (55.2%) were not detected in peripheral blood using the TE-WGS, signifying them as bona fide somatic variants. Further, TE-WGS provided accurate copy number profiles, fusion genes, microsatellite instability, and homologous recombination deficiency scores, which were essential for clinical decision-making. CONCLUSION TE-WGS is a comprehensive approach in personalized oncology, matching TSO500's key biomarker detection capabilities. It uniquely identifies germline variants and genomic instability markers, offering additional clinical actions. Its adaptability and cost-effectiveness underscore its clinical utility, making TE-WGS a valuable tool in personalized cancer treatment.
Collapse
Affiliation(s)
| | - Jin Roh
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Jun Sung Park
- Department of Internal Medicine, Ajou University School of Medicine, Suwon, Korea
| | | | | | | | | | | | | | | | - Ryul Kim
- Inocras Inc., San Diego, CA, USA
| | | | | | | | | | | | - Tae-Hwan Kim
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Yong Won Choi
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Mi Sun Ahn
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Hyun Woo Lee
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
| | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Jang-Hee Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Minsuk Kwon
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, Korea
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Tao XY, Feng SL, Li XJ, Li YJ, Wang W, Gilliham M, Chen ZH, Xu SC. TTLOC: A Tn5 transposase-based approach to localize T-DNA integration sites. PLANT PHYSIOLOGY 2025; 197:kiaf102. [PMID: 40131780 PMCID: PMC11961865 DOI: 10.1093/plphys/kiaf102] [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: 01/05/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025]
Abstract
Thermal asymmetric interlaced-polymerase chain reaction-based and whole-genome sequencing-based T-DNA localization approaches have been developed for the recovery of T-DNA integration sites (TISs). Nevertheless, a low-cost and high-throughput technique for the detection of TISs, which would facilitate the identification of genetically engineered plants, is in high demand for rapid crop breeding and plant synthetic biology. Here, we present Tn5 transposase-based T-DNA integration site localization (TTLOC), a Tn5-based approach for TIS localization. TTLOC employs specialized adaptor-assembled Tn5 transposases for genomic DNA tagmentation. TTLOC library construction is straightforward, involving only six steps that requires two and a half hours to complete. The resulting pooled library is compatible with next-generation sequencing, which enables high-throughput determination. We demonstrate the ability of TTLOC to recover 95 non-redundant TISs from 65 transgenic Arabidopsis (Arabidopsis thaliana) lines, and 37 non-redundant TISs from the genomes of transgenic rice (Oryza sativa), soybean (Glycine max), tomato (Solanum lycopersicum), potato (Solanum tuberosum), and from the large hexaploid wheat (Triticum aestivum) genome. TTLOC is a cost-effective method, as 1 to 2 Gb of raw data for each multiplexing library are sufficient for efficient TIS calling, independent of the genome size. Our results establish TTLOC as a promising strategy for evaluation of genome engineered plants and for selecting genome safe harbors for trait stacking in crop breeding and plant synthetic biology.
Collapse
Affiliation(s)
- Xiao-Yuan Tao
- Biotechnology Institute, Xianghu Laboratory, Hangzhou 311231, China
| | - Shou-Li Feng
- Biotechnology Institute, Xianghu Laboratory, Hangzhou 311231, China
| | - Xin-Jia Li
- Biotechnology Institute, Xianghu Laboratory, Hangzhou 311231, China
| | - Yan-Jun Li
- Biotechnology Institute, Xianghu Laboratory, Hangzhou 311231, China
| | - Wei Wang
- National Key Laboratory of Crop Genetics & Germplasm Enhancement and Utilization, College of Agriculture, Nanjing Agricultural University/Zhongshan Biological Breeding Laboratory/Collaborative Innovation Center for Modern Crop Production (CIC-MCP), Nanjing, Jiangsu 210095, China
| | - Matthew Gilliham
- ARC Centre of Excellence Plants for Space, School of Agriculture, Food and Wine, Waite Research Precinct, University of Adelaide, Glen Osmond, SA 5064, Australia
| | - Zhong-Hua Chen
- School of Science, Western Sydney University, Penrith, NSW 2751, Australia
| | - Sheng-Chun Xu
- Biotechnology Institute, Xianghu Laboratory, Hangzhou 311231, China
- Institute of Digital Agriculture, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| |
Collapse
|
22
|
Yang W, Luyten Y, Reister E, Mangelson H, Sisson Z, Auch B, Liachko I, Roberts R, Ettwiller L. Proxi-RIMS-seq2 applied to native microbiomes uncovers hundreds of known and novel m5C methyltransferase specificities. Nucleic Acids Res 2025; 53:gkaf226. [PMID: 40156868 PMCID: PMC11954522 DOI: 10.1093/nar/gkaf226] [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: 10/12/2024] [Revised: 12/13/2024] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Methylation patterns in bacteria can be used to study restriction-modification or other defense systems with novel properties. While m4C and m6A methylation are well characterized mainly through PacBio sequencing, the landscape of m5C methylation is under-characterized. To bridge this gap, we performed RIMS-seq2 (rapid identification of methyltransferase specificity sequencing) on microbiomes composed of resolved assemblies of distinct genomes through proximity ligation. This high-throughput approach enables the identification of m5C methylated motifs and links them to cognate methyltransferases directly on native microbiomes without the need to isolate bacterial strains. Methylation patterns can also be identified on bacteriophage DNA and compared with host DNA, strengthening evidence for phage-host interactions. Applied to three different microbiomes, the method unveiled over 1900 motifs that were deposited in REBASE. The motifs include a novel eight-base recognition site (CATm5CGATG) that was experimentally validated by characterizing its cognate methyltransferase. Our findings suggest that microbiomes harbor arrays of untapped m5C methyltransferase specificities, providing insights into bacterial biology and biotechnological applications.
Collapse
Affiliation(s)
- Weiwei Yang
- New England Biolabs, Inc., 240 County Road, Ipswich, MA 01938, United States
| | - Yvette Luyten
- New England Biolabs, Inc., 240 County Road, Ipswich, MA 01938, United States
| | - Emily Reister
- Phase Genomics, Inc., 1617 8th Ave N, Seattle, WA 98109, United States
| | - Hayley Mangelson
- Phase Genomics, Inc., 1617 8th Ave N, Seattle, WA 98109, United States
| | - Zach Sisson
- Phase Genomics, Inc., 1617 8th Ave N, Seattle, WA 98109, United States
| | - Benjamin Auch
- Phase Genomics, Inc., 1617 8th Ave N, Seattle, WA 98109, United States
| | - Ivan Liachko
- Phase Genomics, Inc., 1617 8th Ave N, Seattle, WA 98109, United States
| | - Richard J Roberts
- New England Biolabs, Inc., 240 County Road, Ipswich, MA 01938, United States
| | - Laurence Ettwiller
- New England Biolabs, Inc., 240 County Road, Ipswich, MA 01938, United States
| |
Collapse
|
23
|
Khan M, Alteneder M, Reiter W, Krausgruber T, Dobnikar L, Madern M, Waldherr M, Bock C, Hartl M, Ellmeier W, Henriksson J, Boucheron N. Single-cell and chromatin accessibility profiling reveals regulatory programs of pathogenic Th2 cells in allergic asthma. Nat Commun 2025; 16:2565. [PMID: 40089475 PMCID: PMC11910648 DOI: 10.1038/s41467-025-57590-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
Lung pathogenic T helper type 2 (pTh2) cells are important in mediating allergic asthma, but fundamental questions remain regarding their heterogeneity and epigenetic regulation. Here we investigate immune regulation in allergic asthma by single-cell RNA sequencing in mice challenged with house dust mite, in the presence and absence of histone deacetylase 1 (HDAC1) function. Our analyses indicate two distinct highly proinflammatory subsets of lung pTh2 cells and pinpoint thymic stromal lymphopoietin (TSLP) and Tumour Necrosis Factor Receptor Superfamily (TNFRSF) members as important drivers to generate pTh2 cells in vitro. Using our in vitro model, we uncover how signalling via TSLP and a TNFRSF member shapes chromatin accessibility at the type 2 cytokine gene loci by modulating HDAC1 repressive function. In summary, we have generated insights into pTh2 cell biology and establish an in vitro model for investigating pTh2 cells that proves useful for discovering molecular mechanisms involved in pTh2-mediated allergic asthma.
Collapse
Affiliation(s)
- Matarr Khan
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Marlis Alteneder
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Wolfgang Reiter
- Max Perutz Labs, Mass Spectrometry Facility, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Vienna, Austria
| | - Thomas Krausgruber
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Center for Medical Data Science, Institute of Artificial Intelligence, Vienna, Austria
| | - Lina Dobnikar
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Moritz Madern
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Monika Waldherr
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
- FH Campus Wien, Department of Applied Life Sciences/Bioengineering/Bioinformatics, Vienna, Austria
| | - Christoph Bock
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Center for Medical Data Science, Institute of Artificial Intelligence, Vienna, Austria
| | - Markus Hartl
- Max Perutz Labs, Mass Spectrometry Facility, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Center for Molecular Biology, Department of Biochemistry and Cell Biology, Vienna, Austria
| | - Wilfried Ellmeier
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria
| | - Johan Henriksson
- Umeå University, Umeå Centre for Microbial Research (UCMR), Integrated Science Lab (Icelab), Department of Molecular Biology, Umeå, Sweden
| | - Nicole Boucheron
- Medical University of Vienna, Center of Pathophysiology, Infectiology and Immunology, Institute of Immunology, Division of Immunobiology, Vienna, Austria.
| |
Collapse
|
24
|
Gao J, Liu R, Huang K, Li Z, Sheng X, Chakraborty K, Han C, Zhang D, Becker L, Zhao Y. Dynamic investigation of hypoxia-induced L-lactylation. Proc Natl Acad Sci U S A 2025; 122:e2404899122. [PMID: 40030031 PMCID: PMC11912421 DOI: 10.1073/pnas.2404899122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 12/06/2024] [Indexed: 03/19/2025] Open
Abstract
The recently identified histone modification lysine lactylation can be stimulated by L-lactate and glycolysis. Although the chemical group added upon lysine lactylation was originally proposed to be the L-enantiomer of lactate (KL-la), two isomeric modifications, lysine D-lactylation (KD-la) and N-ε-(carboxyethyl) lysine (Kce), also exist in cells, with their precursors being metabolites of glycolysis. The dynamic regulation and differences among these three modifications in response to hypoxia remain poorly understood. In this study, we demonstrate that intracellular KL-la, but not KD-la or Kce, is up-regulated in response to hypoxia. Depletion of glyoxalase enzymes, GLO1 and GLO2, had minimal impact on KD-la, Kce, or hypoxia-induced KL-la. Conversely, blocking glycolytic flux to L-lactate under hypoxic conditions by knocking out lactate dehydrogenase A/B completely abolished the induction of KL-la but increased KD-la and Kce. We further observed a correlation between the level of KL-la and hypoxia-inducible factor 1 alpha (HIF-1α) expression under hypoxic conditions and when small molecules were used to stabilize HIF-1α in the normoxia condition. Our result demonstrated that there is a strong correlation between HIF-1α and KL-la in lung cancer tissues and that patient samples with higher grade tend to have higher KL-la levels. Using a proteomics approach, we quantified 66 KL-la sites that were up-regulated by hypoxia and demonstrated that p300/CBP contributes to hypoxia-induced KL-la. Collectively, our study demonstrates that KL-la, rather than KD-la or Kce, is the prevailing lysine lactylation in response to hypoxia. Our results therefore demonstrate a link between KL-la and the hypoxia-induced adaptation of tumor cells.
Collapse
Affiliation(s)
- Jinjun Gao
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Ruilong Liu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Kevin Huang
- College of Agriculture and Life Science, Cornell University, Ithaca, NY14853
| | - Ziyuan Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI53706
| | - Xinlei Sheng
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Kasturi Chakraborty
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Chang Han
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Di Zhang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing100871, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing100871, China
| | - Lev Becker
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| | - Yingming Zhao
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL60637
| |
Collapse
|
25
|
Thorpe HJ, Pedersen BS, Dietze M, Link N, Quinlan AR, Bonkowsky JL, Thomas A, Chow CY. Identification of CNTN2 as a genetic modifier of PIGA-CDG in a family with incomplete penetrance and in Drosophila. Am J Hum Genet 2025; 112:572-582. [PMID: 39947185 PMCID: PMC11947179 DOI: 10.1016/j.ajhg.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/19/2025] Open
Abstract
Loss-of-function mutations in the X chromosome gene PIGA lead to phosphatidylinositol glycan class A congenital disorder of glycosylation (PIGA-CDG), an ultra-rare CDG typically presenting with seizures, hypotonia, and neurodevelopmental delay. We identified two brothers (probands) with PIGA-CDG, presenting with epilepsy and mild developmental delay. Both probands carry PIGA c.395C>G (p.Ser132Cys), an ultra-rare variant predicted to be damaging. Strikingly, the maternal grandfather and a great uncle also carry the same PIGA variant, but neither presents with symptoms associated with PIGA-CDG. We hypothesized that genetic modifiers might contribute to this reduced penetrance. Using whole-genome sequencing and pedigree analysis, we identified possible susceptibility variants found in the probands and not in the carriers and possible protective variants found in the carriers and not in the probands. Candidate genetic modifier variants included heterozygous, damaging variants in three genes involved directly in glycosylphosphatidylinositol (GPI)-anchor biosynthesis and additional variants in other glycosylation pathways or encoding GPI-anchored proteins. Using a Drosophila eye-based model, we tested modifiers identified through genome sequencing. Loss of CNTN2, a predicted protective modifier that encodes a GPI-anchored protein responsible for neuron/glial interactions, rescues loss of PIGA in the eye-based model, as we predict in the family. Further testing found that the loss of CNTN2 also rescues PIGA-CDG-specific phenotypes, including seizures and climbing defects in Drosophila neurological models of PIGA-CDG. Using pedigree information, genome sequencing, and in vivo testing, we identified CNTN2 as a strong candidate modifier that could explain the incomplete penetrance in this family. Identifying and studying rare disease modifier genes in families may lead to therapeutic targets.
Collapse
Affiliation(s)
- Holly J Thorpe
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Brent S Pedersen
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Miranda Dietze
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Nichole Link
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Aaron R Quinlan
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, USA; Center for Personalized Medicine, Primary Children's Hospital, Salt Lake City, UT, USA
| | - Ashley Thomas
- Department of Neurology, University of Alabama at Birmingham School of Medicine, Birmingham, AL, USA
| | - Clement Y Chow
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
26
|
Foddis M, Blumenau S, Mueller S, Messerschmidt C, Rocca C, Pagnamenta AT, Winek K, Endres M, Meisel A, Tucci A, Bras J, Guerreiro R, Beule D, Dirnagl U, Sassi C. Ide Copy Number Variant Does Not Influence Stroke Severity in 2 C57BL/6J Mouse Models nor in Humans: An Exploratory Study. Stroke 2025; 56:725-736. [PMID: 39866114 PMCID: PMC7617642 DOI: 10.1161/strokeaha.124.049575] [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: 06/29/2024] [Revised: 10/25/2024] [Accepted: 12/05/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND Contrary to the common belief, the most commonly used laboratory C57BL/6J mouse inbred strain presents a distinctive genetic and phenotypic variability, and for several traits, the genotype-phenotype link remains still unknown. Recently, we characterized the most important stroke survival factor such as brain collateral plasticity in 2 brain ischemia C57BL/6J mouse models (bilateral common carotid artery stenosis and middle cerebral artery occlusion) and observed a Mendelian-like fashion of inheritance of the posterior communicating artery (PcomA) patency. Interestingly, a copy number variant (CNV) spanning Ide locus was reported to segregate in an analogous Mendelian-like pattern in the C57BL/6J colonies of the Jackson Laboratory. Given IDE critical role in vascular plasticity, we hypothesized Ide CNV may have explained PcomA variability in C57BL/6J inbred mice. METHODS We applied a combination of techniques (T2-weighted magnetic resonance imaging, time-of-flight angiography, cerebral blood flow imaging, and histology) to characterize the collaterome in 77 C57BL/6J bilateral common carotid artery stenosis, middle cerebral artery occlusion, naive, and sham mice and performed on these Taqman genotyping, exome sequencing, and RNA sequencing. We then investigated the hypothesis that IDE structural variants (CNVs, gain/loss of function mutations) may have influenced the cerebrovascular phenotype in a large cohort of 454 040 cases and controls (UK Biobank, Genomics England). RESULTS We detected an Ide CNV in a bilateral common carotid artery stenosis mouse with 2 patent PcomAs (minor allele frequency, 1.3%), not segregating with the PcomA patency phenotype. In addition, 2 heterozygous IDE CNVs, resulting in loss of function were found in 1 patient with hereditary ataxia, a patient with hereditary congenital heart disease, and 2 healthy individuals (minor allele frequency 9×10-6). Moreover, we report 4 IDE loss of function point mutations (p.Leu5X, p.Met394ValfsX29, p.Pro14SerfsX26, p.Leu889X, minor allele frequency 0.02%) present also in controls or inherited from healthy parents. CONCLUSIONS Ide CNV and loss of function variants are rare, do not crucially influence PcomA variability in C57BL/6J inbred mice, and do not cause a vascular phenotype in humans.
Collapse
Affiliation(s)
- Marco Foddis
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sonja Blumenau
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Susanne Mueller
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Charité-Universitätsmedizin Berlin, NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Berlin, Germany
| | | | - Clarissa Rocca
- Department of Neuromuscular Disease, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | | | - Katarzyna Winek
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Matthias Endres
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Andreas Meisel
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Arianna Tucci
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Jose Bras
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI49503, USA
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI49503, USA
| | - Dieter Beule
- Berlin Institute of Health, BIH, Core Unit Bioinformatics, Berlin, Germany
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Celeste Sassi
- Department of Experimental Neurology, Center for Stroke Research Berlin (CSB), Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
27
|
Zhu G, Rahman CR, Getty V, Odinokov D, Baruah P, Carrié H, Lim AJ, Guo YA, Poh ZW, Sim NL, Abdelmoneim A, Cai Y, Lakshmanan LN, Ho D, Thangaraju S, Poon P, Lau YT, Gan A, Ng S, Koo SL, Chong DQ, Tay B, Tan TJ, Yap YS, Chok AY, Ng MCH, Tan P, Tan D, Wong L, Wong PM, Tan IB, Skanderup AJ. A deep-learning model for quantifying circulating tumour DNA from the density distribution of DNA-fragment lengths. Nat Biomed Eng 2025; 9:307-319. [PMID: 40055581 DOI: 10.1038/s41551-025-01370-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 02/12/2025] [Indexed: 03/21/2025]
Abstract
The quantification of circulating tumour DNA (ctDNA) in blood enables non-invasive surveillance of cancer progression. Here we show that a deep-learning model can accurately quantify ctDNA from the density distribution of cell-free DNA-fragment lengths. We validated the model, which we named 'Fragle', by using low-pass whole-genome-sequencing data from multiple cancer types and healthy control cohorts. In independent cohorts, Fragle outperformed tumour-naive methods, achieving higher accuracy and lower detection limits. We also show that Fragle is compatible with targeted sequencing data. In plasma samples from patients with colorectal cancer, longitudinal analysis with Fragle revealed strong concordance between ctDNA dynamics and treatment responses. In patients with resected lung cancer, Fragle outperformed a tumour-naive gene panel in the prediction of minimal residual disease for risk stratification. The method's versatility, speed and accuracy for ctDNA quantification suggest that it may have broad clinical utility.
Collapse
Affiliation(s)
- Guanhua Zhu
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Centre for Novostics, Hong Kong SAR, China
- Department of Chemical Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chowdhury Rafeed Rahman
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Victor Getty
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Denis Odinokov
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Probhonjon Baruah
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Hanaé Carrié
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- School of Computing, National University of Singapore, Singapore, Singapore
- Institute of Data Science, National University of Singapore, Singapore, Singapore
- Integrative Sciences and Engineering Programme, Graduate School, National University of Singapore, Singapore, Singapore
| | - Avril Joy Lim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Yu Amanda Guo
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Zhong Wee Poh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Ngak Leng Sim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ahmed Abdelmoneim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yutong Cai
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | - Danliang Ho
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Saranya Thangaraju
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Polly Poon
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yi Ting Lau
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Anna Gan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sarah Ng
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Si-Lin Koo
- National Cancer Center Singapore, Singapore, Singapore
| | - Dawn Q Chong
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | - Brenda Tay
- National Cancer Center Singapore, Singapore, Singapore
| | - Tira J Tan
- National Cancer Center Singapore, Singapore, Singapore
| | - Yoon Sim Yap
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | | | - Matthew Chau Hsien Ng
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | - Patrick Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Daniel Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | - Limsoon Wong
- School of Computing, National University of Singapore, Singapore, Singapore
| | - Pui Mun Wong
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Iain Beehuat Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
- National Cancer Center Singapore, Singapore, Singapore
| | - Anders Jacobsen Skanderup
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- School of Computing, National University of Singapore, Singapore, Singapore.
- National Cancer Center Singapore, Singapore, Singapore.
| |
Collapse
|
28
|
Wang P, Mak VC, Rao L, Wu Q, Zhou Y, Sharma R, Kwon SC, Cheung LW. p85β acts as a transcription cofactor and cooperates with BCLAF1 in the nucleus. Nat Commun 2025; 16:2042. [PMID: 40016211 PMCID: PMC11868507 DOI: 10.1038/s41467-025-56532-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 01/16/2025] [Indexed: 03/01/2025] Open
Abstract
p85β is a regulatory subunit of the phosphoinositide 3-kinase (PI3K). Emerging evidence suggests that p85β goes beyond its role in the PI3K and is functional in the nucleus. In this study, we discover that nuclear p85β is enriched at gene loci and regulates gene transcription and that this regulatory role contributes to the oncogenic potential of nuclear p85β. A multi-omics approach reveals the physical interaction and functional cooperativity between nuclear p85β and a transcription factor BCLAF1. We observe genome-wide co-occupancy of p85β and BCLAF1 at gene targets associated with transcriptional responses. Intriguingly, the targetome includes BCLAF1 of which transcription is activated by p85β and BCLAF1, indicating a positive autoregulation. While BCLAF1 recruits p85β to BCLAF1 loci, p85β facilitates the assembly of BCLAF1, the scaffold protein TRIM28 and the zinc finger transcription factor ZNF263, which together act in concert to activate BCLAF1 transcription. Collectively, this study provides functional evidence and mechanistic basis to support a role of nuclear p85β in modulating gene transcription.
Collapse
Affiliation(s)
- Panpan Wang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Victor Cy Mak
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ling Rao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qiuqiu Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yuan Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Rakesh Sharma
- Proteomics and Metabolomics Core, Centre for PanorOmic Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - S Chul Kwon
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lydia Wt Cheung
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
29
|
Macaluso F, Bos T, Chiroli E, Bonaiuti P, Apuan JC, Gross F, Pompei S, Rice LM, Ciliberto A. Evolutionary adaptation to hyperstable microtubules selectively targets tubulins and is empowered by the spindle assembly checkpoint. Cell Rep 2025; 44:115323. [PMID: 39955777 DOI: 10.1016/j.celrep.2025.115323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/15/2024] [Accepted: 01/27/2025] [Indexed: 02/18/2025] Open
Abstract
Microtubules are polymers required for chromosome segregation. Their drug-induced hyperstabilization impairs chromosome segregation and is an established anti-cancer therapy. How cells respond to microtubule hyperstabilization, however, is incompletely understood. To study this, we evolved budding yeast cells expressing a microtubule-hyperstabilizing tubulin mutant and isolated adapted strains. Aneuploidy of specific chromosomes carrying the microtubule regulators STU2 and VIK1/KAR3 was the first observable adaptation. In the longer run, aneuploidies were outcompeted by mutations in α- or β-tubulin, partially overlapping with mutations in cancer patients. Thus, compensation of microtubule hyperstabilization follows a restrained and reproducible path where new mutations combine with the original offending mutation on the same carrier. While partly compensatory, several mutations failed to re-establish fully normal microtubule dynamics. Sustained growth relied on the mitotic checkpoint, indicating that extended mitotic timing limits the genomic instability caused by reduced microtubule dynamics. Our results predict a potential vulnerability of cells resistant to microtubule-hyperstabilizing agents.
Collapse
Affiliation(s)
- Francesca Macaluso
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Tasia Bos
- Departments of Biophysics and Biochemistry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elena Chiroli
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Paolo Bonaiuti
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Jason C Apuan
- Departments of Biophysics and Biochemistry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Fridolin Gross
- ImmunoConcEpT, CNRS UMR5164, Université de Bordeaux, 33076 Bordeaux, France
| | - Simone Pompei
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Luke M Rice
- Departments of Biophysics and Biochemistry, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Andrea Ciliberto
- IFOM-ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy; Pázmány Péter Catholic University, Faculty of Information Technology and Bionics, 1083 Budapest, Hungary.
| |
Collapse
|
30
|
Barcan AS, Humble JL, Kasaragod S, Sajib MSI, Barcan RA, McGinnity P, Welch TJ, Robertson B, Vamanu E, Bacigalupo A, Llewellyn MS, Samsing F. Understanding the transfer and persistence of antimicrobial resistance in aquaculture using a model teleost gut system. Anim Microbiome 2025; 7:18. [PMID: 39987284 PMCID: PMC11846170 DOI: 10.1186/s42523-025-00377-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 01/07/2025] [Indexed: 02/24/2025] Open
Abstract
BACKGROUND The development, progression, and dissemination of antimicrobial resistance (AMR) are determined by interlinked human, animal, and environmental drivers, which pose severe risks to human and livestock health. Conjugative plasmid transfer drives the rapid dissemination of AMR among bacteria. In addition to the judicious use and implementation of stewardship programs, mitigating the spread of antibiotic resistance requires an understanding of the dynamics of AMR transfer among microbial communities, as well as the role of various microbial taxa as potential reservoirs that promote long-term AMR persistence. Here, we employed Hi-C, a high-throughput, culture-free technique, combined with qPCR, to monitor carriage and transfer of a multidrug-resistent (MDR) plasmid within an Atlantic salmon in vitro gut model during florfenicol treatment, a benzenesulfonyl antibiotic widely deployed in fin-fish aquaculture. RESULTS Microbial communities from the pyloric ceaca of three healthy adult farmed salmon were inoculated into three bioreactors simulating the teleost gut, which were developed for the SalmoSim gut system. The model system was then inoculated with the Escherichia coli strain ATCC 25922 carrying the plasmid pM07-1 and treated with florfenicol at a concentration of 150 mg/L in fish feed media for 5 days prior to the washout/recovery phase. Hi-C and metagenomic sequencing identified numerous transfer events, including those involving gram-negative and gram-positive taxa, and, crucially, the transfer and persistence of the plasmid continued once florfenicol treatment was withdrawn. CONCLUSIONS Our findings highlight the role of the commensal teleost gut flora as a reservoir for AMR even once antimicrobial selective pressure has been withdrawn. Our system also provides a model to study how different treatment regimens and interventions may be deployed to mitigate AMR persistence.
Collapse
Affiliation(s)
- Alexandru S Barcan
- SBOHVM, University of Glasgow, Graham Kerr Building, Glasgow, G12 8QQ, UK
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464, Bucharest, Romania
| | - Joseph L Humble
- SBOHVM, University of Glasgow, Graham Kerr Building, Glasgow, G12 8QQ, UK
| | - Sandeep Kasaragod
- SBOHVM, University of Glasgow, Graham Kerr Building, Glasgow, G12 8QQ, UK
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | | | - Rares A Barcan
- Maths & Physical Sciences, University of Sussex, Falmer, Brighton, BN1 9RH, UK
| | - Philip McGinnity
- School of Biological, Earth and Environmental Sciences, University College Cork, Cork, Ireland
| | - Timothy J Welch
- U.S. Department of Agriculture/Agricultural Research Service, National Center for Cool and Cold Water Aquaculture, Leetown, WV, 25430, USA
| | - Brendan Robertson
- SBOHVM, University of Glasgow, Graham Kerr Building, Glasgow, G12 8QQ, UK
| | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464, Bucharest, Romania
| | | | - Martin S Llewellyn
- SBOHVM, University of Glasgow, Graham Kerr Building, Glasgow, G12 8QQ, UK.
| | - Francisca Samsing
- Sydney School of Veterinary Science, The University of Sydney, Camden, NSW, 2570, Australia
| |
Collapse
|
31
|
Fu H, Huang K, Zhu W, Zhang L, Bandaru R, Wang L, Liu Y, Xia Z. Circulating cell-free DNA methylation profiles as noninvasive multiple sclerosis biomarkers: A proof-of-concept study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.14.25322180. [PMID: 40034794 PMCID: PMC11875267 DOI: 10.1101/2025.02.14.25322180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
In multiple sclerosis (MS), there is a critical need for non-invasive biomarkers to concurrently classify disease subtypes, evaluate disability severity, and predict long-term progression. In this proof-of-concept study, we performed low-coverage whole-genome bisulfite sequencing (WGBS) on 75 plasma cell-free DNA (cfDNA) samples and assessed the clinical utility of cfDNA methylation as a single assay for distinguishing MS patients from non-MS controls, identifying MS subtypes, estimating disability severity, and predicting disease trajectories. We identified thousands of differentially methylated CpGs and hundreds of differentially methylated regions (DMRs) that significantly distinguished MS from controls, separated MS subtypes, and stratified disability severity levels. These DMRs were highly enriched in immunologically and neurologically relevant regulatory elements (e.g., active promoters and enhancers) and contained motifs associated with neuronal function and T-cell differentiation. To distinguish MS subtypes and severity groups, we achieved area-under-the-curve (AUC) values ranging from 0.67 to 0.81 using DMRs and 0.70 to 0.82 using inferred tissue-of-origin patterns from cfDNA methylation, significantly outperforming benchmark neurofilament light chain (NfL) and glial fibrillary acidic protein (GFAP) in the same cohort. Finally, a linear mixed-effects model identified "prognostic regions" where baseline cfDNA methylation levels were associated with disease progression and predicted future disability severity (AUC=0.81) within a 4-year evaluation window. As we plan to generate higher-depth WGBS data and validation in independent cohorts, the present findings suggest the potential clinical utility of circulating cfDNA methylation profiles as promising noninvasive biomarkers in MS diagnosis and prognosis.
Collapse
Affiliation(s)
- Hailu Fu
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Kevin Huang
- Computational Sciences, gRED, Genentech Inc. South San Francisco, CA 94080
| | - Wen Zhu
- University of Pittsburgh, Pittsburgh, PA 15260
| | - Lili Zhang
- University of Pittsburgh, Pittsburgh, PA 15260
| | - Ravi Bandaru
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Li Wang
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Yaping Liu
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL 60611
| | - Zongqi Xia
- University of Pittsburgh, Pittsburgh, PA 15260
| |
Collapse
|
32
|
Azam S, Sahu A, Pandey NK, Neupane M, Van Tassell CP, Rosen BD, Gandham RK, Rath SN, Majumdar SS. Advancing the Indian cattle pangenome: characterizing non-reference sequences in Bos indicus. J Anim Sci Biotechnol 2025; 16:21. [PMID: 39915889 PMCID: PMC11804092 DOI: 10.1186/s40104-024-01133-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/26/2024] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND India harbors the world's largest cattle population, encompassing over 50 distinct Bos indicus breeds. This rich genetic diversity underscores the inadequacy of a single reference genome to fully capture the genomic landscape of Indian cattle. To comprehensively characterize the genomic variation within Bos indicus and, specifically, dairy breeds, we aim to identify non-reference sequences and construct a comprehensive pangenome. RESULTS Five representative genomes of prominent dairy breeds, including Gir, Kankrej, Tharparkar, Sahiwal, and Red Sindhi, were sequenced using 10X Genomics 'linked-read' technology. Assemblies generated from these linked-reads ranged from 2.70 Gb to 2.77 Gb, comparable to the Bos indicus Brahman reference genome. A pangenome of Bos indicus cattle was constructed by comparing the newly assembled genomes with the reference using alignment and graph-based methods, revealing 8 Mb and 17.7 Mb of novel sequence respectively. A confident set of 6,844 Non-reference Unique Insertions (NUIs) spanning 7.57 Mb was identified through both methods, representing the pangenome of Indian Bos indicus breeds. Comparative analysis with previously published pangenomes unveiled 2.8 Mb (37%) commonality with the Chinese indicine pangenome and only 1% commonality with the Bos taurus pangenome. Among these, 2,312 NUIs encompassing ~ 2 Mb, were commonly found in 98 samples of the 5 breeds and designated as Bos indicus Common Insertions (BICIs) in the population. Furthermore, 926 BICIs were identified within 682 protein-coding genes, 54 long non-coding RNAs (lncRNA), and 18 pseudogenes. These protein-coding genes were enriched for functions such as chemical synaptic transmission, cell junction organization, cell-cell adhesion, and cell morphogenesis. The protein-coding genes were found in various prominent quantitative trait locus (QTL) regions, suggesting potential roles of BICIs in traits related to milk production, reproduction, exterior, health, meat, and carcass. Notably, 63.21% of the bases within the BICIs call set contained interspersed repeats, predominantly Long Interspersed Nuclear Elements (LINEs). Additionally, 70.28% of BICIs are shared with other domesticated and wild species, highlighting their evolutionary significance. CONCLUSIONS This is the first report unveiling a robust set of NUIs defining the pangenome of Bos indicus breeds of India. The analyses contribute valuable insights into the genomic landscape of desi cattle breeds.
Collapse
Affiliation(s)
- Sarwar Azam
- National Institute of Animal Biotechnology, Hyderabad, India
- Indian Institute of Technology Hyderabad, Sangareddy, India
| | - Abhisek Sahu
- National Institute of Animal Biotechnology, Hyderabad, India
| | | | - Mahesh Neupane
- Animal Genomics and Improvement Laboratory, USDA-ARS, Beltsville, MD, 20705, USA
| | - Curtis P Van Tassell
- Animal Genomics and Improvement Laboratory, USDA-ARS, Beltsville, MD, 20705, USA
| | - Benjamin D Rosen
- Animal Genomics and Improvement Laboratory, USDA-ARS, Beltsville, MD, 20705, USA.
| | - Ravi Kumar Gandham
- National Institute of Animal Biotechnology, Hyderabad, India.
- Animal Biotechnology, ICAR-NBAGR, Karnal, Haryana, India.
| | | | | |
Collapse
|
33
|
Corbett RJ, Kaufman RS, McQuaid SW, Vaksman Z, Phul S, Brown MA, Mason JL, Waszak SM, Zhang B, Zhong C, Desai H, Hausler R, Naqvi AS, Chroni A, Geng Z, Gonzalez EM, Zhu Y, Heath AP, Li M, Storm PB, Resnick AC, Maxwell KN, Cole KA, Waanders AJ, Bornhorst M, MacFarland SP, Rokita JL, Diskin SJ. Germline pathogenic variation impacts somatic alterations and patient outcomes in pediatric CNS tumors. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.04.25321499. [PMID: 39974082 PMCID: PMC11838646 DOI: 10.1101/2025.02.04.25321499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The contribution of rare pathogenic/likely pathogenic (P/LP) germline variants to pediatric central nervous system (CNS) tumor development remains understudied. Here, we characterized the prevalence and clinical significance of germline P/LP variants in cancer predisposition genes across 830 CNS tumor patients from the Pediatric Brain Tumor Atlas (PBTA). We identified germline P/LP variants in 24.2% (201/830) of patients and the majority (154/201) lacked clinical reporting of genetic tumor syndromes. Among P/LP carriers, 30.7% had putative somatic second hits or loss of function tumor alterations. Finally, we linked pathogenic germline variation with novel somatic events and patient survival to highlight the impact of germline variation on tumorigenesis and patient outcomes.
Collapse
Affiliation(s)
- Ryan J Corbett
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rebecca S Kaufman
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shelly W McQuaid
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zalman Vaksman
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Saksham Phul
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Miguel A Brown
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jennifer L Mason
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sebastian M Waszak
- Laboratory of Computational Neuro-Oncology, Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Bo Zhang
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chuwei Zhong
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Heena Desai
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan Hausler
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ammar S Naqvi
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Antonia Chroni
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Zhuangzhuang Geng
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Elizabeth M Gonzalez
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Yuankun Zhu
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Allison P Heath
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marilyn Li
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Phillip B Storm
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Adam C Resnick
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kara N Maxwell
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina A Cole
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Angela J Waanders
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Miriam Bornhorst
- Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Suzanne P MacFarland
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jo Lynne Rokita
- Center for Data-Driven Discovery in Biomedicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Neurosurgery, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, USA
- Department of Pediatrics, The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Sharon J Diskin
- Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Navasca A, Singh J, Rivera-Varas V, Gill U, Secor G, Baldwin T. Dispensable genome and segmental duplications drive the genome plasticity in Fusarium solani. FRONTIERS IN FUNGAL BIOLOGY 2025; 6:1432339. [PMID: 39974207 PMCID: PMC11835900 DOI: 10.3389/ffunb.2025.1432339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025]
Abstract
Fusarium solani is a species complex encompassing a large phylogenetic clade with diverse members occupying varied habitats. We recently reported a unique opportunistic F. solani associated with unusual dark galls in sugarbeet. We assembled the chromosome-level genome of the F. solani sugarbeet isolate strain SB1 using Oxford Nanopore and Hi-C sequencing. The average size of F. solani genomes is 54 Mb, whereas SB1 has a larger genome of 59.38 Mb, organized into 15 chromosomes. The genome expansion of strain SB1 is due to the high repeats and segmental duplications within its three potentially accessory chromosomes. These chromosomes are absent in the closest reference genome with chromosome-level assembly, F. vanettenii 77-13-4. Segmental duplications were found in three chromosomes but are most extensive between two specific SB1 chromosomes, suggesting that this isolate may have doubled its accessory genes. Further comparison of the F. solani strain SB1 genome demonstrates inversions and syntenic regions to an accessory chromosome of F. vanettenii 77-13-4. The pan-genome of 12 publicly available F. solani isolates nearly reached gene saturation, with few new genes discovered after the addition of the last genome. Based on orthogroups and average nucleotide identity, F. solani is not grouped by lifestyle or origin. The pan-genome analysis further revealed the enrichment of several enzymes-coding genes within the dispensable (accessory + unique genes) genome, such as hydrolases, transferases, oxidoreductases, lyases, ligases, isomerase, and dehydrogenase. The evidence presented here suggests that genome plasticity, genetic diversity, and adaptive traits in Fusarium solani are driven by the dispensable genome with significant contributions from segmental duplications.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas Baldwin
- Department of Plant Pathology, North Dakota State University, Fargo, ND, United States
| |
Collapse
|
35
|
La Torre R, Hamilton JP, Saucedo-Bazalar M, Caycho E, Vaillancourt B, Wood JC, Ramírez M, Buell CR, Orjeda G. A chromosome-level genome assembly of the Peruvian Algarrobo (Neltuma pallida) provides insights on its adaptation to its unique ecological niche. G3 (BETHESDA, MD.) 2025; 15:jkae283. [PMID: 39657049 PMCID: PMC11797065 DOI: 10.1093/g3journal/jkae283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/10/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024]
Abstract
The dry forests of northern Peru are dominated by the legumous tree Neltuma pallida which is adapted to hot arid and semiarid conditions in the tropics. Despite having been successfully introduced in multiple other areas around the world, N. pallida is currently threatened in its native area, where it is invaluable for the dry forest ecosystem and human subsistence. A major tool for enhancing ecosystem conservation and understanding the adaptive properties of N. pallida to dry forest ecosystems is the construction of a reference genome sequence. Here, we report on a high-quality reference genome for N. pallida. The final genome assembly size is 403.7 Mb, consisting of 14 pseudochromosomes and 63 scaffolds with an N50 size of 26.2 Mb and a 34.3% GC content. Use of Benchmarking Universal Single Copy Orthologs revealed 99.2% complete orthologs. Long terminal repeat elements dominated the repetitive sequence content which was 51.2%. Genes were annotated using N. pallida transcripts, plant protein sequences, and ab initio predictions resulting in 22,409 protein-coding genes encoding 24,607 gene models. Comparative genomic analysis showed evidence of rapidly evolving gene families related to disease resistance, transcription factors, and signaling pathways. The chromosome-scale N. pallida reference genome will be a useful resource for understanding plant evolution in extreme and highly variable environments.
Collapse
Affiliation(s)
- Renato La Torre
- Laboratory of Genomics and Bioinformatics for Biodiversity, Faculty of Biological Sciences, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| | - John P Hamilton
- Center for Applied Genetic Technologies, University of Georgia, Athens, GA 30602, USA
- Department of Crop and Soil Sciences, University of Georgia, Athens, GA 30602, USA
| | - Manuel Saucedo-Bazalar
- Laboratory of Molecular Biology, Department of Biology and Biochemistry, Universidad Nacional de Tumbes, Tumbes 24001, Peru
| | - Esteban Caycho
- Laboratory of Genomics and Bioinformatics for Biodiversity, Faculty of Biological Sciences, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| | - Brieanne Vaillancourt
- Center for Applied Genetic Technologies, University of Georgia, Athens, GA 30602, USA
| | - Joshua C Wood
- Center for Applied Genetic Technologies, University of Georgia, Athens, GA 30602, USA
| | - Manuel Ramírez
- Laboratory of Genomics and Bioinformatics for Biodiversity, Faculty of Biological Sciences, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| | - C Robin Buell
- Center for Applied Genetic Technologies, University of Georgia, Athens, GA 30602, USA
- Department of Crop and Soil Sciences, University of Georgia, Athens, GA 30602, USA
- Institute of Plant Breeding, Genetics, and Genomics, University of Georgia, Athens, GA 30602, USA
- The Plant Center, University of Georgia, Athens, GA 30602, USA
| | - Gisella Orjeda
- Laboratory of Genomics and Bioinformatics for Biodiversity, Faculty of Biological Sciences, Universidad Nacional Mayor de San Marcos, Lima 15081, Peru
| |
Collapse
|
36
|
Liu R, Ren X, Park YE, Feng H, Sheng X, Song X, AminiTabrizi R, Shah H, Li L, Zhang Y, Abdullah KG, Dubois-Coyne S, Lin H, Cole PA, DeBerardinis RJ, McBrayer SK, Huang H, Zhao Y. Nuclear GTPSCS functions as a lactyl-CoA synthetase to promote histone lactylation and gliomagenesis. Cell Metab 2025; 37:377-394.e9. [PMID: 39642882 PMCID: PMC11798710 DOI: 10.1016/j.cmet.2024.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/18/2024] [Accepted: 11/09/2024] [Indexed: 12/09/2024]
Abstract
Histone lysine lactylation is a physiologically and pathologically relevant epigenetic pathway that can be stimulated by the Warburg effect-associated L-lactate. Nevertheless, the mechanism by which cells use L-lactate to generate lactyl-coenzyme A (CoA) and how this process is regulated remains unknown. Here, we report the identification of guanosine triphosphate (GTP)-specific SCS (GTPSCS) as a lactyl-CoA synthetase in the nucleus. The mechanism was elucidated through the crystallographic structure of GTPSCS in complex with L-lactate, followed by mutagenesis experiments. GTPSCS translocates into the nucleus and interacts with p300 to elevate histone lactylation but not succinylation. This process depends on a nuclear localization signal in the GTPSCS G1 subunit and acetylation at G2 subunit residue K73, which mediates the interaction with p300. GTPSCS/p300 collaboration synergistically regulates histone H3K18la and GDF15 expression, promoting glioma proliferation and radioresistance. GTPSCS represents the inaugural enzyme to catalyze lactyl-CoA synthesis for epigenetic histone lactylation and regulate oncogenic gene expression in glioma.
Collapse
Affiliation(s)
- Ruilong Liu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Xuelian Ren
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Yae Eun Park
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Huixu Feng
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinlei Sheng
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaohan Song
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Roya AminiTabrizi
- Biological Science Division, Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Hardik Shah
- Biological Science Division, Metabolomics Platform, Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA
| | - Lingting Li
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Yu Zhang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai 200032, China
| | - Kalil G Abdullah
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Hillman Comprehensive Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15232, USA
| | - Sarah Dubois-Coyne
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Hening Lin
- Howard Hughes Medical Institute, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Philip A Cole
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Ralph J DeBerardinis
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Samuel K McBrayer
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - He Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - Yingming Zhao
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Comprehensive Cancer Center, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
37
|
Castellano KR, Neitzey ML, Starovoitov A, Barrett GA, Reid NM, Vuruputoor VS, Webster CN, Storer JM, Pauloski NR, Ameral NJ, McEvoy SL, McManus MC, Puritz JB, Wegrzyn JL, O’Neill RJ. Genome Assembly of a Living Fossil, the Atlantic Horseshoe Crab Limulus polyphemus, Reveals Lineage-Specific Whole-Genome Duplications, Transposable Element-Based Centromeres, and a ZW Sex Chromosome System. Mol Biol Evol 2025; 42:msaf021. [PMID: 39907027 PMCID: PMC11836539 DOI: 10.1093/molbev/msaf021] [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: 09/13/2024] [Revised: 12/16/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Horseshoe crabs, considered living fossils with a stable morphotype spanning ∼445 million years, are evolutionarily, ecologically, and biomedically important species experiencing rapid population decline. Of the four extant species of horseshoe crabs, the Atlantic horseshoe crab, Limulus polyphemus, has become an essential component of the modern medicine toolkit. Here, we present the first chromosome-level genome assembly, and the most contiguous and complete assembly to date, for L. polyphemus using nanopore long-read sequencing and chromatin conformation analysis. We find support for three horseshoe crab-specific whole-genome duplications, but none shared with Arachnopulmonata (spiders and scorpions). Moreover, we discovered tandem duplicates of endotoxin detection pathway components Factors C and G, identify candidate centromeres consisting of Gypsy retroelements, and classify the ZW sex chromosome system for this species and a sister taxon, Carcinoscorpius rotundicauda. Finally, we revealed this species has been experiencing a steep population decline over the last 5 million years, highlighting the need for international conservation interventions and fisheries-based management for this critical species.
Collapse
Affiliation(s)
- Kate R Castellano
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Michelle L Neitzey
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Andrew Starovoitov
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT 06269, USA
| | - Gabriel A Barrett
- Biological and Environmental Sciences, University of Rhode Island, Kingston, RI 02881, USA
| | - Noah M Reid
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Vidya S Vuruputoor
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT 06269, USA
| | - Cynthia N Webster
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT 06269, USA
| | - Jessica M Storer
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Nicole R Pauloski
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
| | - Natalie J Ameral
- Biological and Environmental Sciences, University of Rhode Island, Kingston, RI 02881, USA
- Division of Marine Fisheries, Rhode Island Department of Environmental Management, Providence, RI 02908, USA
| | - Susan L McEvoy
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT 06269, USA
| | - M Conor McManus
- Division of Marine Fisheries, Rhode Island Department of Environmental Management, Providence, RI 02908, USA
| | - Jonathan B Puritz
- Department of Biological Sciences, University of Rhode Island, Kingston, RI 02881, USA
| | - Jill L Wegrzyn
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT 06269, USA
| | - Rachel J O’Neill
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269, USA
- Institute for Systems Genomics, University of Connecticut, Storrs, CT 06269, USA
- Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| |
Collapse
|
38
|
Han SY, Cho SH, Jung SH, Kang M, Suh MW, Park MK, Lee JH, Lee SY. Genotypes and clinical phenotypes of pediatric patients with NOG variants: Middle ear surgical outcomes from a Tertiary Center in South Korea. Int J Pediatr Otorhinolaryngol 2025; 189:112230. [PMID: 39837070 DOI: 10.1016/j.ijporl.2025.112230] [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: 10/31/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
OBJECTIVE Although NOG variants are linked to congenital stapes fixation and conductive hearing loss (CHL), little is known about middle ear surgery outcomes and the characteristics of accompanying inner ear anomalies. We explored auditory phenotypes in patients with NOG variants, with a focus on the outcomes of middle ear surgery. METHODS This study included 11 patients from five unrelated Korean families harboring NOG variants. Genomic investigations were conducted using whole-exome sequencing and whole-genome sequencing. The clinical phenotypes, including pre- and postoperative audiological profiles, radiological abnormalities, and other comorbidities, were analyzed. RESULTS The average age at genetic testing was 8.2 years (range, 0-13 years). Two previously reported NOG variants (c.509C > T:p.Pro170Leu and c.252dup:p.Glu85ArgfsTer97) and three novel NOG variants, including the c.187G > T: p.Glu63Ter and two cryptic large deletion within the 17q22.2 region, were identified. All patients exhibited non-progressive CHL. Inner ear anomalies were documented in two patients, with variations such as cochlea and vestibular dysplasia. In this study, seven ears of four patients underwent stapedotomy, resulting in a significantly reduced air-bone gap of 10.18 ± 1.48 dB (P = 0.016), with sustained improvement. Conversely, patients carrying p.Pro170Leu variant, which is associated with poor outcomes for middle ear surgery, were excluded from surgical consideration. CONCLUSION We expanded the spectrum of genotypes and auditory phenotypes associated with NOG variants. Surgical intervention for CHL underlying NOG variants elicits favorable outcomes. However, clinicians should consider the potential for poor prognosis in certain NOG variants. Collectively, identifying NOG variants could guide the treatment strategies to improve CHL.
Collapse
Affiliation(s)
- Sang-Yoon Han
- Department of Otolaryngology-Head and Neck Surgery, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Sung Ho Cho
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Sung Ho Jung
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Myeongsin Kang
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Myung-Whan Suh
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea
| | - Moo Kyun Park
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea
| | - Jun Ho Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea
| | - Sang-Yeon Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Sensory Organ Research Institute, Seoul National University, Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Panzeri I, Fagnocchi L, Apostle S, Tompkins M, Wolfrum E, Madaj Z, Hostetter G, Liu Y, Schaefer K, Yang CH, Bergsma A, Drougard A, Dror E, Chandler DP, Schramek D, Triche TJ, Pospisilik JA. TRIM28-dependent developmental heterogeneity determines cancer susceptibility through distinct epigenetic states. NATURE CANCER 2025; 6:385-403. [PMID: 39856421 PMCID: PMC11864977 DOI: 10.1038/s43018-024-00900-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 12/13/2024] [Indexed: 01/27/2025]
Abstract
Mutations in cancer risk genes increase susceptibility, but not all carriers develop cancer. Indeed, while DNA mutations are necessary drivers of cancer, only a small subset of mutated cells go on to cause the disease. To date, the mechanisms underlying individual cancer susceptibility remain unclear. Here, we took advantage of a unique mouse model of intrinsic developmental heterogeneity (Trim28+/D9) to investigate whether early-life epigenetic variation influences cancer susceptibility later in life. We found that heterozygosity of Trim28 is sufficient to generate two distinct early-life epigenetic states associated with differing cancer susceptibility. These developmentally primed states exhibit differential methylation patterns at typically silenced heterochromatin, detectable as early as 10 days of age. The differentially methylated loci are enriched for genes with known oncogenic potential, frequently mutated in human cancers and correlated with poor prognosis. This study provides genetic evidence that intrinsic developmental heterogeneity can prime individual, lifelong cancer susceptibility.
Collapse
Affiliation(s)
- Ilaria Panzeri
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA.
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
| | - Luca Fagnocchi
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Stefanos Apostle
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Megan Tompkins
- Vivarium and Transgenics Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Emily Wolfrum
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Zachary Madaj
- Bioinformatics and Biostatistics Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Galen Hostetter
- Pathology and Biorepository Core, Van Andel Institute, Grand Rapids, MI, USA
| | - Yanqing Liu
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Kristen Schaefer
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Chih-Hsiang Yang
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alexis Bergsma
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
- Parkinson's Disease Center, Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, USA
| | - Anne Drougard
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Erez Dror
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | | | - Daniel Schramek
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Timothy J Triche
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
- Department of Pediatrics, MSU College of Human Medicine, East Lansing, MI, USA
- Department of Translational Genomics, University of Southern California, Los Angeles, CA, USA
| | - John Andrew Pospisilik
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA.
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
| |
Collapse
|
40
|
Lan L, Li H, Xu S, Xu Y, Leng Q, Zhang L, Wu L, Yin J, Wu Z, Niu J. Chromosome-level and haplotype-resolved genome assembly of Bougainvillea glabra. Sci Data 2025; 12:107. [PMID: 39827170 PMCID: PMC11742905 DOI: 10.1038/s41597-024-04333-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Bougainvillea glabra Choisy, a common perennial shrub or climbing vine in tropical and subtropical areas, has significant ornamental and ecological value due to its showy floriferous blooms and adaptability to multiple environmental stresses. In this study, based on PacBio and Hi-C sequencing data, we presented the haplotype-resolved, and chromosome-level assembled genomes for B. glabra (2n = 34). We obtained two haplotypes with genome size of 2,855,692,602 bp and 2,889,511,499 bp, N50 of 165 Mbp and 167 Mbp, and both anchored to 17 chromosomes with anchor ratio of 97.07% and 97.55%, respectively. Through the BUSCO evaluation process, the two assembled haplotypes both exhibited the high score of 96%. By combining the high-confidence different evidence of genome structure annotation pipeline we annotated each haplotype containing ~33,000 predicted genes, of which 86% were functionally annotated by different databases. These high-quality haplotypes assembly and annotation results will serve as a great genomic resource to explore the ornamental traits and environmental adaptability and accelerate improvement in future.
Collapse
Affiliation(s)
- Lan Lan
- National Key Laboratory for Tropical Crop Breeding / Key Laboratory of Gene Resources and Germplasm Enhancement in Southern China, MARA / Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- State Agricultural Biotechnology Centre (SABC), College of Science, Health, Engineering and Education, Murdoch University, Perth, 6150, WA, Australia
| | - Haiyan Li
- National Key Laboratory for Tropical Crop Breeding / Key Laboratory of Gene Resources and Germplasm Enhancement in Southern China, MARA / Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- The Engineering Technology Research Center of Tropical Ornamental Plant Germplasm Innovation and Utilization, Danzhou, China
| | - Shisong Xu
- National Key Laboratory for Tropical Crop Breeding / Key Laboratory of Gene Resources and Germplasm Enhancement in Southern China, MARA / Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- The Engineering Technology Research Center of Tropical Ornamental Plant Germplasm Innovation and Utilization, Danzhou, China
| | - Yueting Xu
- National Key Laboratory for Tropical Crop Breeding / Key Laboratory of Gene Resources and Germplasm Enhancement in Southern China, MARA / Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- Graduate School of Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Qingyun Leng
- National Key Laboratory for Tropical Crop Breeding / Key Laboratory of Gene Resources and Germplasm Enhancement in Southern China, MARA / Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- The Engineering Technology Research Center of Tropical Ornamental Plant Germplasm Innovation and Utilization, Danzhou, China
| | - Linbi Zhang
- Hainan Daxin Landscape Co., Ltd, Haikou, China
| | - Linqiao Wu
- Hainan Big Lake Bridge Technology Co., Ltd, Haikou, China
| | - Junmei Yin
- National Key Laboratory for Tropical Crop Breeding / Key Laboratory of Gene Resources and Germplasm Enhancement in Southern China, MARA / Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
- The Engineering Technology Research Center of Tropical Ornamental Plant Germplasm Innovation and Utilization, Danzhou, China
- Sanya Research Institute, Chinese Academy of Tropical Agricultural Sciences, Sanya, China
| | - Zhiqiang Wu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China.
| | - Junhai Niu
- National Key Laboratory for Tropical Crop Breeding / Key Laboratory of Gene Resources and Germplasm Enhancement in Southern China, MARA / Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China.
- The Engineering Technology Research Center of Tropical Ornamental Plant Germplasm Innovation and Utilization, Danzhou, China.
| |
Collapse
|
41
|
Liu Y, Hrit JA, Chomiak AA, Stransky S, Hoffman JR, Tiedemann RL, Wiseman AK, Kariapper LS, Dickson BM, Worden EJ, Fry CJ, Sidoli S, Rothbart SB. DNA hypomethylation promotes UHRF1-and SUV39H1/H2-dependent crosstalk between H3K18ub and H3K9me3 to reinforce heterochromatin states. Mol Cell 2025; 85:394-412.e12. [PMID: 39631394 PMCID: PMC11741932 DOI: 10.1016/j.molcel.2024.11.009] [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: 03/20/2024] [Revised: 09/17/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024]
Abstract
Mono-ubiquitination of lysine 18 on histone H3 (H3K18ub), catalyzed by UHRF1, is a DNMT1 docking site that facilitates replication-coupled DNA methylation maintenance. Its functions beyond this are unknown. Here, we genomically map simultaneous increases in UHRF1-dependent H3K18ub and SUV39H1/H2-dependent H3K9me3 following DNMT1 inhibition. Mechanistically, transient accumulation of hemi-methylated DNA at CpG islands facilitates UHRF1 recruitment and E3 ligase activity toward H3K18. Notably, H3K18ub enhances SUV39H1/H2 methyltransferase activity and, in colon cancer cells, nucleates new H3K9me3 domains at CpG island promoters of DNA methylation-silenced tumor suppressor genes (TSGs). Disrupting UHRF1 enzyme activity prevents H3K9me3 accumulation while promoting PRC2-dependent H3K27me3 as a tertiary layer of gene repression in these regions. By contrast, disrupting H3K18ub-dependent SUV39H1/H2 activity enhances the transcriptional activating and antiproliferative effects of DNMT1 inhibition. Collectively, these findings reveal roles for UHRF1 and H3K18ub in regulating a hierarchy of repressive histone methylation signaling and rationalize a combination strategy for epigenetic cancer therapy.
Collapse
Affiliation(s)
- Yanqing Liu
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Joel A Hrit
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Alison A Chomiak
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Stephanie Stransky
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | - Ashley K Wiseman
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Leena S Kariapper
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Bradley M Dickson
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA
| | - Evan J Worden
- Department of Structural Biology, Van Andel Institute, Grand Rapids, MI 49503, USA
| | | | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Scott B Rothbart
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI 49503, USA.
| |
Collapse
|
42
|
Paulo DF, Nguyen TNM, Ward CM, Corpuz RL, Kauwe AN, Rendon P, Ruano REY, Cardoso AAS, Gouvi G, Fung E, Crisp P, Okada A, Choo A, Stauffer C, Bourtzis K, Sim SB, Baxter SW, Geib SM. Functional genomics implicates ebony in the black pupae phenotype of tephritid fruit flies. Commun Biol 2025; 8:60. [PMID: 39814836 PMCID: PMC11736145 DOI: 10.1038/s42003-025-07489-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
The remarkable diversity of insect pigmentation offers a captivating avenue for studying evolution and genetics. In tephritids, understanding the molecular basis of mutant traits is also crucial for applied entomology, enabling the creation of genetic sexing strains through genome editing, thus facilitating sex-sorting before sterile insect releases. Here, we present evidence from classical and modern genetics showing that the black pupae (bp) phenotype in the GUA10 strain of Anastrepha ludens is caused by a large deletion at the ebony locus, removing the gene's entire coding region. Targeted knockout of ebony induced analogous bp phenotypes across six major tephritid agricultural pests, demonstrating that disruption of Ebony alone is sufficient to produce the mutant trait in distantly related species. This functional characterization further allowed a deeper exploration of Ebony's role in pigmentation and development across life stages in diverse species. Our findings offer key insights for molecular engineering of sexing strains based on the bp marker and for future evolutionary developmental biology studies in tephritids.
Collapse
Affiliation(s)
- Daniel F Paulo
- Department of Plant and Environmental Protection Sciences, University of Hawai'i at Mānoa, Honolulu, USA
- U.S. Department of Agriculture, Agriculture Research Service, Daniel K. Inouye U.S. Pacific Basin Agricultural Research Center, Hilo, USA
| | - Thu N M Nguyen
- School of BioSciences, The University of Melbourne, Melbourne, Australia
| | - Chris M Ward
- School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - Renee L Corpuz
- U.S. Department of Agriculture, Agriculture Research Service, Daniel K. Inouye U.S. Pacific Basin Agricultural Research Center, Hilo, USA
| | - Angela N Kauwe
- U.S. Department of Agriculture, Agriculture Research Service, Daniel K. Inouye U.S. Pacific Basin Agricultural Research Center, Hilo, USA
| | - Pedro Rendon
- International Atomic Energy Agency, Technical Cooperation, Division for Latin America and the Caribbean, MOSCAMED Program, Guatemala City, Guatemala
| | | | - Amanda A S Cardoso
- Insect Pest Control Laboratory, Department of Nuclear Sciences and Applications, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Seibersdorf, Austria
| | - Georgia Gouvi
- Insect Pest Control Laboratory, Department of Nuclear Sciences and Applications, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Seibersdorf, Austria
| | - Elisabeth Fung
- School of Agriculture, Food and Wine, University of Adelaide, Adelaide, Australia
- South Australian Research and Development Institute, Urrbrae, Australia
| | - Peter Crisp
- School of Agriculture, Food and Wine, University of Adelaide, Adelaide, Australia
- South Australian Research and Development Institute, Urrbrae, Australia
| | - Anzu Okada
- School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - Amanda Choo
- School of Biological Sciences, The University of Adelaide, Adelaide, Australia
| | - Christian Stauffer
- Department of Ecosystem Management, Climate and Biodiversity, Boku University, Vienna, Austria
| | - Kostas Bourtzis
- Insect Pest Control Laboratory, Department of Nuclear Sciences and Applications, Joint FAO/IAEA Centre of Nuclear Techniques in Food and Agriculture, Seibersdorf, Austria
| | - Sheina B Sim
- U.S. Department of Agriculture, Agriculture Research Service, Daniel K. Inouye U.S. Pacific Basin Agricultural Research Center, Hilo, USA
| | - Simon W Baxter
- School of BioSciences, The University of Melbourne, Melbourne, Australia.
| | - Scott M Geib
- U.S. Department of Agriculture, Agriculture Research Service, Daniel K. Inouye U.S. Pacific Basin Agricultural Research Center, Hilo, USA.
| |
Collapse
|
43
|
Sen S, Estève PO, Raman K, Beaulieu J, Chin H, Feehery G, Vishnu U, Xu SY, Samuelson J, Pradhan S. Distinct structural and functional heterochromatin partitioning of lamin B1 and lamin B2 revealed using genome-wide nicking enzyme epitope targeted DNA sequencing. Nucleic Acids Res 2025; 53:gkae1317. [PMID: 39817518 PMCID: PMC11736435 DOI: 10.1093/nar/gkae1317] [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: 07/18/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 01/18/2025] Open
Abstract
Gene expression is regulated by chromatin DNA methylation and other features, including histone post-translational modifications (PTMs), chromatin remodelers and transcription factor occupancy. A complete understanding of gene regulation will require the mapping of these chromatin features in small cell number samples. Here we describe a novel genome-wide chromatin profiling technology, named as Nicking Enzyme Epitope targeted DNA sequencing (NEED-seq). NEED-seq offers antibody-targeted controlled nicking by Nt.CviPII-pGL fusion to study specific protein-DNA complexes in formaldehyde fixed cells, allowing for both visual and genomic resolution of epitope bound chromatin. When applied to nuclei, NEED-seq yielded genome-wide profile of chromatin-associated proteins and histone PTMs. Additionally, NEED-seq of lamin B1 and B2 demonstrated their association with heterochromatin. Lamin B1- and B2-associated domains (LAD) segregated to three different states, and states with stronger LAD correlated with heterochromatic marks. Hi-C analysis displayed A and B compartment with equal lamin B1 and B2 distribution, although methylated DNA remained high in B compartment. LAD clustering with Hi-C resulted in subcompartments, with lamin B1 and B2 partitioning to facultative and constitutive heterochromatin, respectively, and were associated with neuronal development. Thus, lamin B1 and B2 show structural and functional partitioning in mammalian nucleus.
Collapse
Affiliation(s)
- Sagnik Sen
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Pierre-Olivier Estève
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Karthikeyan Raman
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Julie Beaulieu
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Hang Gyeong Chin
- PMBBRC, Gyeongsang National University, Jinju 52828, Republic of Korea
| | - George R Feehery
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | | | - Shuang-yong Xu
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - James C Samuelson
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| | - Sriharsa Pradhan
- Molecular Genetics and Genomics, New England Biolabs, Inc, 240 County Road, Ipswich, MA 01938, USA
| |
Collapse
|
44
|
Blanco E, Camps C, Bahal S, Kerai MD, Ferla MP, Rochussen AM, Handel AE, Golwala ZM, Spiridou Goncalves H, Kricke S, Klein F, Zhang F, Zinghirino F, Evans G, Keane TM, Lizot S, Kusters MA, Iro MA, Patel SV, Morris EC, Burns SO, Radcliffe R, Vasudevan P, Price A, Gillham O, Valdebenito GE, Stewart GS, Worth A, Adams SP, Duchen M, André I, Adams DJ, Santili G, Gilmour KC, Holländer GA, Davies EG, Taylor JC, Griffiths GM, Thrasher AJ, Dhalla F, Kreins AY. Dominant negative variants in ITPR3 impair T cell Ca2+ dynamics causing combined immunodeficiency. J Exp Med 2025; 222:e20220979. [PMID: 39560673 PMCID: PMC11577440 DOI: 10.1084/jem.20220979] [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: 06/06/2022] [Revised: 09/09/2024] [Accepted: 10/25/2024] [Indexed: 11/20/2024] Open
Abstract
The importance of calcium (Ca2+) as a second messenger in T cell signaling is exemplified by genetic deficiencies of STIM1 and ORAI1, which abolish store-operated Ca2+ entry (SOCE) resulting in combined immunodeficiency (CID). We report five unrelated patients with de novo missense variants in ITPR3, encoding a subunit of the inositol 1,4,5-trisphosphate receptor (IP3R), which forms a Ca2+ channel in the endoplasmic reticulum (ER) membrane responsible for the release of ER Ca2+ required to trigger SOCE, and for Ca2+ transfer to other organelles. The patients presented with CID, abnormal T cell Ca2+ homeostasis, incompletely penetrant ectodermal dysplasia, and multisystem disease. Their predominant T cell immunodeficiency is characterized by significant T cell lymphopenia, defects in late stages of thymic T cell development, and impaired function of peripheral T cells, including inadequate NF-κB- and NFAT-mediated, proliferative, and metabolic responses to activation. Pathogenicity is not due to haploinsufficiency, rather ITPR3 protein variants interfere with IP3R channel function leading to depletion of ER Ca2+ stores and blunted SOCE in T cells.
Collapse
Affiliation(s)
- Elena Blanco
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Carme Camps
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Sameer Bahal
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Mohit D. Kerai
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Immunology Laboratory, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Matteo P. Ferla
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Adam M. Rochussen
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Adam E. Handel
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Zainab M. Golwala
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Helena Spiridou Goncalves
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Susanne Kricke
- SIHMDS-Haematology Laboratory, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Fabian Klein
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
| | - Fang Zhang
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Federica Zinghirino
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Grace Evans
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Thomas M. Keane
- Wellcome Sanger Institute, Cambridge, UK
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK
| | - Sabrina Lizot
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
| | - Maaike A.A. Kusters
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Mildred A. Iro
- Department of Paediatric Infectious Diseases and Immunology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Faculty of Medicine and Institute of Life Sciences, University of Southampton, Southampton, UK
| | - Sanjay V. Patel
- Department of Paediatric Infectious Diseases and Immunology, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Emma C. Morris
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK
- Institute for Immunity and Transplantation, University College London, London, UK
| | - Siobhan O. Burns
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, UK
- Institute for Immunity and Transplantation, University College London, London, UK
| | - Ruth Radcliffe
- Department of Immunology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Pradeep Vasudevan
- Department of Clinical Genetics, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Arthur Price
- Department of Immunology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Olivia Gillham
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, University College London, London, UK
| | - Gabriel E. Valdebenito
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, University College London, London, UK
| | - Grant S. Stewart
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Austen Worth
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Stuart P. Adams
- SIHMDS-Haematology Laboratory, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Michael Duchen
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, University College London, London, UK
| | - Isabelle André
- Human Lymphohematopoiesis Laboratory, Imagine Institute, INSERM UMR 1163, Université Paris Cité, Paris, France
| | | | - Giorgia Santili
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Kimberly C. Gilmour
- Immunology Laboratory, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Georg A. Holländer
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Paediatric Immunology, Department of Biomedicine, University of Basel and University Children’s Hospital, Basel, Switzerland
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - E. Graham Davies
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Jenny C. Taylor
- National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Gillian M. Griffiths
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Adrian J. Thrasher
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Fatima Dhalla
- Department of Paediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Clinical Immunology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Alexandra Y. Kreins
- Molecular and Cellular Immunology, Great Ormond Street Institute of Child Health, University College London, London, UK
- Department of Paediatric Immunology and Gene Therapy, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, UK
| |
Collapse
|
45
|
Hovhannisyan A, Delser PM, Hakobyan A, Jones ER, Schraiber JG, Antonosyan M, Margaryan A, Xue Z, Jeon S, Bhak J, Hrechdakian P, Sahakyan H, Saag L, Khachatryan Z, Yepiskoposyan L, Manica A. Demographic history and genetic variation of the Armenian population. Am J Hum Genet 2025; 112:11-27. [PMID: 39591962 PMCID: PMC11739871 DOI: 10.1016/j.ajhg.2024.10.022] [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: 03/01/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
We introduce a sizable (n = 34) whole-genome dataset on Armenians, a population inhabiting the region in West Asia known as the Armenian highlands. Equipped with this genetic data, we conducted a whole-genome study of Armenians and deciphered their fine-scale population structure and complex demographic history. We demonstrated that the Armenian populations from western, central, and eastern parts of the highlands are relatively homogeneous. The Sasun, a population in the south that had been argued to have received a major genetic contribution from Assyrians, was instead shown to have derived its slightly divergent genetic profile from a bottleneck that occurred in the recent past. We also investigated the debated question on the genetic origin of Armenians and failed to find any significant support for historical suggestions by Herodotus of their Balkan-related ancestry. We checked the degree of continuity of modern Armenians with ancient inhabitants of the eastern Armenian highlands and detected a genetic input into the region from a source linked to Neolithic Levantine Farmers at some point after the Early Bronze Age. Additionally, we cataloged an abundance of new mutations unique to the population, including a missense mutation predicted to cause familial Mediterranean fever, an autoinflammatory disorder highly prevalent in Armenians. Thus, we highlight the importance of further genetic and medical studies of this population.
Collapse
Affiliation(s)
- Anahit Hovhannisyan
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin D02 PN40, Ireland; Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK; Institute of Molecular Biology, National Academy of Sciences, Yerevan 0014, Armenia; Estonian Biocentre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia.
| | - Pierpaolo Maisano Delser
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin D02 PN40, Ireland; Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Anna Hakobyan
- Institute of Molecular Biology, National Academy of Sciences, Yerevan 0014, Armenia; Department of Structural and Computational Biology, Max Perutz Labs, University of Vienna, 1030 Vienna, Austria
| | - Eppie R Jones
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin D02 PN40, Ireland; Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Joshua G Schraiber
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90018, USA
| | - Mariya Antonosyan
- Institute of Molecular Biology, National Academy of Sciences, Yerevan 0014, Armenia; Max Planck Institute of Geoanthropology, 07745 Jena, Germany
| | - Ashot Margaryan
- Institute of Molecular Biology, National Academy of Sciences, Yerevan 0014, Armenia; Centre for GeoGenetics, Natural History Museum of Denmark, University of Copenhagen, 1350 Copenhagen, Denmark
| | - Zhe Xue
- Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Sungwon Jeon
- Korean Genomics Center (KOGIC), Ulsan National Institute of Science and Technology (UNIST), Uslan 44919, Republic of Korea; Clinomics Inc., Ulsan 44919, Republic of Korea
| | - Jong Bhak
- Korean Genomics Center (KOGIC), Ulsan National Institute of Science and Technology (UNIST), Uslan 44919, Republic of Korea; Clinomics Inc., Ulsan 44919, Republic of Korea
| | | | - Hovhannes Sahakyan
- Institute of Molecular Biology, National Academy of Sciences, Yerevan 0014, Armenia; Estonian Biocentre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Lehti Saag
- Estonian Biocentre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Zaruhi Khachatryan
- Institute of Molecular Biology, National Academy of Sciences, Yerevan 0014, Armenia
| | - Levon Yepiskoposyan
- Institute of Molecular Biology, National Academy of Sciences, Yerevan 0014, Armenia.
| | - Andrea Manica
- Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| |
Collapse
|
46
|
Schafran P, Hauser DA, Nelson JM, Xu X, Mueller LA, Kulshrestha S, Smalley I, de Vries S, Irisarri I, de Vries J, Davies K, Villarreal JCA, Li FW. Pan-phylum genomes of hornworts reveal conserved autosomes but dynamic accessory and sex chromosomes. NATURE PLANTS 2025; 11:49-62. [PMID: 39753957 DOI: 10.1038/s41477-024-01883-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 11/27/2024] [Indexed: 01/25/2025]
Abstract
Hornworts, one of the three bryophyte phyla, show some of the deepest divergences in extant land plants, with some families separated by more than 300 million years. Previous hornwort genomes represented only one genus, limiting the ability to infer evolution within hornworts and their early land plant ancestors. Here we report ten new chromosome-scale genomes representing all hornwort families and most of the genera. We found that, despite the deep divergence, synteny was surprisingly conserved across all hornwort genomes, a pattern that might be related to the absence of whole-genome duplication. We further uncovered multiple accessory and putative sex chromosomes that are highly repetitive and CpG methylated. In contrast to autosomes, these chromosomes mostly lack syntenic relationships with one another and are evolutionarily labile. Notable gene retention and losses were identified, including those responsible for flavonoid biosynthesis, stomata patterning and phytohormone reception, which have implications in reconstructing the evolution of early land plants. Together, our pan-phylum genomes revealed an array of conserved and divergent genomic features in hornworts, highlighting the uniqueness of this deeply diverged lineage.
Collapse
Affiliation(s)
| | | | | | - Xia Xu
- Boyce Thompson Institute, Ithaca, NY, USA
| | - Lukas A Mueller
- Boyce Thompson Institute, Ithaca, NY, USA
- Plant Breeding and Genetics Section, Cornell University, Ithaca, NY, USA
| | - Samarth Kulshrestha
- The New Zealand Institute for Plant and Food Research Limited, Palmerston North, New Zealand
| | - Isabel Smalley
- Department of Biology, University of Minnesota Duluth, Duluth, MN, USA
| | - Sophie de Vries
- Institute of Microbiology and Genetics, Göttingen Center for Molecular Biosciences, Campus Institute Data Science, Department of Applied Bioinformatics, University of Göttingen, Göttingen, Germany
| | - Iker Irisarri
- Institute of Microbiology and Genetics, Göttingen Center for Molecular Biosciences, Campus Institute Data Science, Department of Applied Bioinformatics, University of Göttingen, Göttingen, Germany
- Section Phylogenomics, Centre for Molecular Biodiversity Research, Leibniz Institute for the Analysis of Biodiversity Change, Zoological Museum Hamburg, Hamburg, Germany
| | - Jan de Vries
- Institute of Microbiology and Genetics, Göttingen Center for Molecular Biosciences, Campus Institute Data Science, Department of Applied Bioinformatics, University of Göttingen, Göttingen, Germany
| | - Kevin Davies
- The New Zealand Institute for Plant and Food Research Limited, Palmerston North, New Zealand
| | | | - Fay-Wei Li
- Boyce Thompson Institute, Ithaca, NY, USA.
- Plant Biology Section, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
47
|
McCorison CB, Kim T, Donato JJ, LaPara TM. Proximity-Ligation Metagenomic Sequence Analysis Reveals That the Antibiotic Resistome Makes Significant Transitions During Municipal Wastewater Treatment. Environ Microbiol 2025; 27:e70036. [PMID: 39797441 PMCID: PMC11724201 DOI: 10.1111/1462-2920.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/20/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025]
Abstract
Shotgun and proximity-ligation metagenomic sequencing were used to generate thousands of metagenome assembled genomes (MAGs) from the untreated wastewater, activated sludge bioreactors, and anaerobic digesters from two full-scale municipal wastewater treatment facilities. Analysis of the antibiotic resistance genes (ARGs) in the pool of contigs from the shotgun metagenomic sequences revealed significantly different relative abundances and types of ARGs in the untreated wastewaster compared to the activated sludge bioreactors or the anaerobic digesters (p < 0.05). In contrast, these results were statistically similar when comparing the ARGs in the pool of MAGs, suggesting that proximity-ligation metagenomic sequencing is particularly useful for pairing ARGs with their hosts but less adept at discerning quantitative differences in ARG types and relative abundances. For example, numerous MAGs of the genera Acinetobacter, Enterococcus, Klebsiella and Pseudomonas were identified in the untreated wastewater, many of which harboured plasmid-borne and/or chromosomal-borne ARGs; none of these MAGs, however, were detected in the activated sludge bioreactors or anaerobic digesters. In conclusion, this research demonstrates that the antibiotic resistome undergoes significant transitions in both the relative abundance and the host organisms during the municipal wastewater treatment process.
Collapse
Affiliation(s)
| | - Taegyu Kim
- Department of Genetics and Genomic SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Justin J. Donato
- Department of ChemistryUniversity of St. ThomasSt. PaulMinnesotaUSA
| | - Timothy M. LaPara
- Department of Civil, Environmental, and Geo‐EngineeringUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
48
|
Wang N, Pachai MR, Li D, Lee CJ, Warda S, Khudoynazarova MN, Cho WH, Xie G, Shah SR, Yao L, Qian C, Wong EWP, Yan J, Tomas FV, Hu W, Kuo F, Gao SP, Luo J, Smith AE, Han M, Gao D, Ge K, Yu H, Chandarlapaty S, Iyer GV, Rosenberg JE, Solit DB, Al-Ahmadie HA, Chi P, Chen Y. Loss of Kmt2c or Kmt2d primes urothelium for tumorigenesis and redistributes KMT2A-menin to bivalent promoters. Nat Genet 2025; 57:165-179. [PMID: 39806204 PMCID: PMC11735410 DOI: 10.1038/s41588-024-02015-y] [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: 06/20/2023] [Accepted: 10/23/2024] [Indexed: 01/16/2025]
Abstract
Members of the KMT2C/D-KDM6A complex are recurrently mutated in urothelial carcinoma and in histologically normal urothelium. Here, using genetically engineered mouse models, we demonstrate that Kmt2c/d knockout in the urothelium led to impaired differentiation, augmented responses to growth and inflammatory stimuli and sensitization to oncogenic transformation by carcinogen and oncogenes. Mechanistically, KMT2D localized to active enhancers and CpG-poor promoters that preferentially regulate the urothelial lineage program and Kmt2c/d knockout led to diminished H3K4me1, H3K27ac and nascent RNA transcription at these sites, which leads to impaired differentiation. Kmt2c/d knockout further led to KMT2A-menin redistribution from KMT2D localized enhancers to CpG-high and bivalent promoters, resulting in derepression of signal-induced immediate early genes. Therapeutically, Kmt2c/d knockout upregulated epidermal growth factor receptor signaling and conferred vulnerability to epidermal growth factor receptor inhibitors. Together, our data posit that functional loss of Kmt2c/d licenses a molecular 'field effect' priming histologically normal urothelium for oncogenic transformation and presents therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Naitao Wang
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohini R Pachai
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dan Li
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Cindy J Lee
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarah Warda
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Woo Hyun Cho
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Guojia Xie
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sagar R Shah
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Li Yao
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Cheng Qian
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elissa W P Wong
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juan Yan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fanny V Tomas
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wenhuo Hu
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fengshen Kuo
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sizhi P Gao
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jiaqian Luo
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alison E Smith
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ming Han
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Dong Gao
- State Key Laboratory of Cell Biology, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Kai Ge
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Haiyuan Yu
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Gopakumar V Iyer
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Jonathan E Rosenberg
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - David B Solit
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Hikmat A Al-Ahmadie
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ping Chi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
49
|
Manning LK, Eager KLM, Willet CE, Slattery S, McNally JH, Spiers ZB, Hazelton M, Child G, Duggan R, O’Rourke BA, Tammen I. Myotonia Congenita in Australian Merino Sheep with a Missense Variant in CLCN1. Animals (Basel) 2024; 14:3703. [PMID: 39765607 PMCID: PMC11672719 DOI: 10.3390/ani14243703] [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: 11/09/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Myotonia congenita is a hereditary, non-dystrophic skeletal muscle disorder associated with muscle stiffness due to delayed muscle relaxation after contraction. We review myotonia congenita in domesticated animals and humans and investigated suspected myotonia congenita in a flock of Merino sheep in Australia. In 2020, a property in New South Wales reported a four-year history of lambs that would fall on disturbance before rapidly recovering, with 13 affected sheep identified in 2020. Episodes were associated with a short period of tetanic spasms and a stiff gait upon rising. Lambs were otherwise normal between episodes, although over time, lost body condition and occasionally died from misadventure. An inherited condition was considered from limited pedigree information and a preliminary diagnosis of myotonia congenita was made based on clinical presentation. Biochemistry from four sheep found variable, but typically mild increases in creatine kinase (CK) and aspartate aminotransferase (AST). Modified electromyography on six affected sheep found irregular electrical activity within the muscle. For four sheep, there were no consistent significant abnormalities on post mortem examination and histopathology-typical for this condition. A review of the Online Mendelian Inheritance in Man (OMIM) and Online Mendelian Inheritance in Animals (OMIA) databases was conducted to summarise information about myotonia congenita in humans and eight non-human species of animals. Comparing the characteristic clinical presentation, pathology and electromyography data of affected Merino sheep to similar conditions in other species assisted the identification of likely candidate genes. Whole genome sequencing of two affected lambs detected a missense variant in CLCN1 (NC_056057.1:g.107930611C>T; XM_004008136.5:c.844C>T; XP_004008185.4:p.(P282S)), with a predicted deleterious effect on protein function. An SNP genotyping assay was developed, and the variant segregated with the disease in 12 affected sheep and obligate carrier rams under an assumed recessive mode of inheritance. Identifying a likely causal variant and developing a diagnostic test allows screening of suspected affected or carrier Merino sheep for early intervention to reduce propagation of the variant within flocks.
Collapse
Affiliation(s)
- Leah K. Manning
- Sydney School of Veterinary Science, The University of Sydney, Camden, NSW 2570, Australia
- Elizabeth Macarthur Agricultural Institute, Department of Primary Industries and Regional Development, Woodbridge Road, Menangle, NSW 2568, Australia
| | - Katie L. M. Eager
- Sydney School of Veterinary Science, The University of Sydney, Camden, NSW 2570, Australia
- Elizabeth Macarthur Agricultural Institute, Department of Primary Industries and Regional Development, Woodbridge Road, Menangle, NSW 2568, Australia
| | - Cali E. Willet
- Sydney Informatics Hub, Core Research Facilities, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Shaun Slattery
- North West Local Land Services, Narrabri, NSW 2390, Australia
| | | | - Zoe B. Spiers
- Elizabeth Macarthur Agricultural Institute, Department of Primary Industries and Regional Development, Woodbridge Road, Menangle, NSW 2568, Australia
| | - Mark Hazelton
- Elizabeth Macarthur Agricultural Institute, Department of Primary Industries and Regional Development, Woodbridge Road, Menangle, NSW 2568, Australia
| | - Georgina Child
- Small Animal Specialist Hospital, North Ryde, NSW 2113, Australia
| | - Rick Duggan
- North West Local Land Services, Narrabri, NSW 2390, Australia
| | - Brendon A. O’Rourke
- Elizabeth Macarthur Agricultural Institute, Department of Primary Industries and Regional Development, Woodbridge Road, Menangle, NSW 2568, Australia
| | - Imke Tammen
- Sydney School of Veterinary Science, The University of Sydney, Camden, NSW 2570, Australia
| |
Collapse
|
50
|
Ren P, Zhang M, Khan MZ, Yang L, Jing Y, Liu X, Yang X, Zhang C, Zhang M, Zhu Z, Zheng N, Zhang L, Zhang S, Zhu M. Genome-Wide Structural Variation Analysis and Breed Comparison of Local Domestic Ducks in Shandong Province, China. Animals (Basel) 2024; 14:3657. [PMID: 39765561 PMCID: PMC11672513 DOI: 10.3390/ani14243657] [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: 11/08/2024] [Revised: 11/27/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Structural variations in the duck genome significantly impact the environmental adaptability and phenotypic diversity of duck populations. Characterizing these SVs in local domestic duck breeds from Shandong province offers valuable insights for breed selection and the development of new breeds. This study aimed to profile the genomic SVs in three local duck breeds (Matahu duck, Weishan partridge duck, and Wendeng black duck) and explore their differential distributions. A total of 21,673 SVs were detected using LUMPY (v0.2.13) and DELLY (v1.0.3) software, with 46% located in intergenic regions, 33% in intronic regions, and frameshift deletions being the most prevalent in exonic regions (3%). SVs distribution showed a decreasing trend with shorter chromosome lengths. Population structure analysis revealed distinct genetic profiles, with Matahu and Weishan partridge ducks showing closer affinities and the Wendeng black duck having a more homogeneous genetic background, likely due to geographic isolation. Functional annotation identified genes related to nervous system development, mitosis, spindle assembly, and energy metabolism. Notable genes included PLXNA4, NRP2, SEMA3A, PTEN, MYBL2, ADK, and COX4I1. Additionally, genes such as PRKG1, GABRA2, and FSHR were linked to energy metabolism and reproductive activity. The study provides a comprehensive analysis of SVs, revealing significant genetic differentiation and identifying genes associated with economically important traits, offering valuable resources for the genetic improvement and breeding of local duck breeds.
Collapse
Affiliation(s)
- Pengwei Ren
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| | - Meixia Zhang
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| | - Muhammad Zahoor Khan
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| | - Liu Yang
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| | - Yadi Jing
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| | - Xiang Liu
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| | - Xiaohui Yang
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| | - Chaoran Zhang
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| | - Min Zhang
- Shandong Animal Husbandry Station, Jinan 250010, China
| | - Zhiming Zhu
- Fujian Key Laboratory of Animal Genetics and Breeding, Institute of Animal Husbandry and Veterinary Medicine of Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - Nenzhu Zheng
- Fujian Key Laboratory of Animal Genetics and Breeding, Institute of Animal Husbandry and Veterinary Medicine of Fujian Academy of Agricultural Sciences, Fuzhou 350013, China
| | - Lujiao Zhang
- Weihai Wendeng District Animal Husbandry and Veterinary Career Development Center, Weihai 264400, China
| | - Shuer Zhang
- Shandong Animal Husbandry Station, Jinan 250010, China
| | - Mingxia Zhu
- College of Agriculture and Biology, Liaocheng University, Liaocheng 252000, China
| |
Collapse
|