1
|
Grimsley HE, Antczak M, Reddin IG, Weiler N, McLaughlin KM, Rothweiler F, Haas J, Nist A, Mernberger M, Stiewe T, Fenton TR, Speidel D, Harper-Wynne C, Cox K, Heckl D, Cinatl J, Wass MN, Garrett MD, Michaelis M. Using a novel panel of drug-resistant triple-negative breast cancer cell lines to identify candidate therapeutic targets and biomarkers. Cancer Lett 2025; 624:217754. [PMID: 40300663 DOI: 10.1016/j.canlet.2025.217754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 04/16/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Here, we introduce a novel set of triple-negative breast cancer (TNBC) cell lines consisting of MDA-MB-468, HCC38, and HCC1806 and their sublines adapted to cisplatin, doxorubicin, eribulin, paclitaxel, gemcitabine, or 5-fluorouracil. Whole exome sequencing combined with TCGA-derived patient data resulted in the identification of 682 biomarker candidates in a pan-cancer analysis. Thirty-five genes were considered the most promising candidates because they harbored resistance-associated variants in at least two resistant sublines, and their expression correlated with TNBC patient survival. Exome sequencing and response profiles to cytotoxic drugs and DNA damage response inhibitors identified revealed remarkably little overlap between the resistant sublines, suggesting that each resistance formation process follows a unique route. This reflects recent findings on cancer cell evolution in patients, supporting the relevance of drug-adapted cancer cell lines as preclinical models of acquired resistance. Moreover, all of the drug-resistant TNBC sublines remained sensitive or even displayed collateral sensitivity to a range of tested compounds. Cross-resistance levels were lowest for the CHK2 inhibitor CCT241533, the PLK1 inhibitor SBE13, and the RAD51 recombinase inhibitor B02, suggesting that CHK2, PLK1, and RAD51 are potential drug targets for therapy-refractory TNBC. In conclusion, we present novel preclinical models of acquired drug resistance in TNBC and the identification of novel candidate therapeutic targets and biomarkers for this disease.
Collapse
Affiliation(s)
- Helen E Grimsley
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK; Department of Radiation Oncology and the Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Magdalena Antczak
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| | - Ian G Reddin
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK; Bio-R Bioinformatics Research Facility, Faculty of Medicine, University of Southampton, UK
| | - Nicole Weiler
- Dr Petra Joh-Research Institute, 60528, Frankfurt am Main, Germany; Fachbereich Biowissenschaften der Goethe-Universität, 60438, Frankfurt am Main, Germany
| | - Katie-May McLaughlin
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| | | | - Johannes Haas
- Department of Pediatrics, Goethe University Frankfurt, 60590, Frankfurt am Main, Germany; Institute for Experimental Pediatric Hematology and Oncology (EPHO), 60528, Frankfurt am Main, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, 35043, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35032, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, 35043, Marburg, Germany; Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35032, Marburg, Germany
| | - Tim R Fenton
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Daniel Speidel
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, New South Wales, Australia
| | - Catherine Harper-Wynne
- Kent Oncology Centre, Maidstone and Tunbridge Wells NHS Trust, Hermitage Lane, Maidstone, ME16 9QQ, UK
| | - Karina Cox
- Peggy Wood Breast Unit, Maidstone Hospital, Hermitage Lane, Maidstone, Kent, ME16 9QQ, UK
| | - Dirk Heckl
- Department of Pediatrics, Goethe University Frankfurt, 60590, Frankfurt am Main, Germany; Institute for Experimental Pediatric Hematology and Oncology (EPHO), 60528, Frankfurt am Main, Germany
| | - Jindrich Cinatl
- Dr Petra Joh-Research Institute, 60528, Frankfurt am Main, Germany.
| | - Mark N Wass
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK.
| | - Michelle D Garrett
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK.
| | - Martin Michaelis
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK; Dr Petra Joh-Research Institute, 60528, Frankfurt am Main, Germany.
| |
Collapse
|
2
|
Nogimura D, Moriyasu K, Ishida S, Kohda M, Yazawa T, Morita M. SNP associations in the L-citrulline metabolic pathway and vascular aging in the Japanese population. PLoS One 2025; 20:e0323778. [PMID: 40440257 PMCID: PMC12121766 DOI: 10.1371/journal.pone.0323778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/15/2025] [Indexed: 06/02/2025] Open
Abstract
Decreased nitric oxide (NO) production from the vascular endothelium is a major factor for vascular aging. Because vascular aging has few specific subjective symptoms, assessing the susceptibility to vascular aging is beneficial for its early detection and improvement. Therefore, this study evaluated the associations between single nucleotide polymorphisms (SNPs) on L-citrulline (Cit) pathways essential for NO production and the health characteristics involved in vascular aging using a candidate gene approach. Associations with a significance level included those between the KCNMB4 rs17108108 C allele and tendency to gain weight, the ADCY8 rs6470860 G allele and numbness of limbs, the NOS1 rs2271987 T allele and lower back pain, and the PDE9A rs2284972 G allele and body pain with negative mood states. A genome-wide association study was also conducted to analyze SNPs more extensively across genes related to Cit and NO metabolism, which revealed genome-wide significant associations between the PRMT6 rs12028323 C allele and mood disturbance. These significant associations could be explained by the change in downstream NO signaling, supporting the relationship between the investigated traits and vascular function. These traits can be manifested as subjective symptoms of vascular aging. Therefore, the identified SNPs could predict susceptibility to the subjective symptoms of vascular aging, which could lead to genotype-based personalized interventions of Cit for an efficient improvement of vascular health.
Collapse
Affiliation(s)
- Dai Nogimura
- Institute of Health Sciences, Kirin Holdings Company, Ltd., Fujisawa, Kanagawa, Japan
| | - Kazuki Moriyasu
- Institute of Health Sciences, Kirin Holdings Company, Ltd., Fujisawa, Kanagawa, Japan
| | | | | | | | - Masahiko Morita
- Institute of Health Sciences, Kirin Holdings Company, Ltd., Fujisawa, Kanagawa, Japan
| |
Collapse
|
3
|
Jimenez NR, Herman CR, Łaniewski P, Cope E, Lee K, Mahnert ND, Chase DM, Caporaso JG, Herbst-Kralovetz MM. Navigating complexities of polymorphic microbiomes in endometrial cancer. NPJ Biofilms Microbiomes 2025; 11:85. [PMID: 40404643 PMCID: PMC12098703 DOI: 10.1038/s41522-025-00690-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 04/01/2025] [Indexed: 05/24/2025] Open
Abstract
The microbiome is key to understanding endometrial cancer (EC) etiology and prevention strategies, implicated in the regulation of estrogen in estrogen-driven cancers. Utilizing robust methodologies in the QIIME 2 platform, we examined 16S rRNA vaginal and rectal microbiome data from an EC cohort: 192 women with benign gynecologic conditions, endometrial hyperplasia, or endometrial cancer. Distinct microbial compositions and community networks specific to EC were identified and related to histological grade with adjustments for EC risk factors. Vaginal health-associated Lactobacillus and Limosilactobacillus, and rectal Prevotella and Peptoniphilus, were depleted in EC, while detrimental vaginal Anaerococcus, Porphyromonas, Prevotella, Peptoniphilus, and rectal Buttiaxella were enriched. Significant bacterial features were shared between rectal and vaginal sites in EC, such as Prevotella timonensis and Peptoniphilus A. Vaginal Lactobacillus abundance contributed to less feature sharing from the rectum. Putative microbial metabolic analysis identified dysregulation of amino acid, complex carbohydrate, and hormone metabolism amongst patients with EC.
Collapse
Affiliation(s)
- Nicole R Jimenez
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Chloe R Herman
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
- School of Informatics, Computing and Cyber Systems, Northern Arizona University, Flagstaff, AZ, USA
| | - Paweł Łaniewski
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Emily Cope
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
| | - Keehoon Lee
- Translational Genomics Research Institute, part of City of Hope, Flagstaff, AZ, USA
| | - Nichole D Mahnert
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Dana M Chase
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - J Gregory Caporaso
- Center for Applied Microbiome Science, Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA.
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
| | - Melissa M Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
- Department of Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
| |
Collapse
|
4
|
Lin MJ, Langmead B, Safonova Y. IGLoo enables comprehensive analysis and assembly of immunoglobulin heavy-chain loci in lymphoblastoid cell lines using PacBio high-fidelity reads. CELL REPORTS METHODS 2025; 5:101033. [PMID: 40315852 DOI: 10.1016/j.crmeth.2025.101033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/04/2025]
Abstract
High-quality human genome assemblies derived from lymphoblastoid cell lines (LCLs) provide reference genomes and pangenomes for genomics studies. However, LCLs pose technical challenges for profiling immunoglobulin (IG) genes, as their IG loci contain a mixture of germline and somatically recombined haplotypes, making genotyping and assembly difficult with widely used frameworks. To address this, we introduce IGLoo, a software tool that analyzes sequence data and assemblies derived from LCLs, characterizing somatic V(D)J recombination events and identifying breakpoints and missing IG genes in the assemblies. Furthermore, IGLoo implements a reassembly framework to improve germline assembly quality by integrating information on somatic events and population structural variations in IG loci. Applying IGLoo to the assemblies from the Human Pangenome Reference Consortium, we gained valuable insights into the mechanisms, gene usage, and patterns of V(D)J recombination and the causes of assembly artifacts in the IG heavy-chain (IGH) locus, and we improved the representation of IGH assemblies.
Collapse
Affiliation(s)
- Mao-Jan Lin
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Ben Langmead
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Yana Safonova
- Department of Computer Science, Johns Hopkins University, Baltimore, MD 21218, USA; Computer Science and Engineering Department, Pennsylvania State University, University Park, PA 16802, USA; Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
5
|
Sarhan MS, Antonello G, Weissensteiner H, Mengoni C, Mascalzoni D, Waldron L, Segata N, Fuchsberger C. Human mitochondrial DNA in public metagenomes: Opportunity or privacy threat? Cell 2025; 188:2561-2566. [PMID: 40378832 DOI: 10.1016/j.cell.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/03/2025] [Accepted: 03/11/2025] [Indexed: 05/19/2025]
Abstract
Human DNA is unavoidably present in metagenomic analyses of human microbiomes. While current protocols remove human DNA before submission to public repositories, mitochondrial DNA (mtDNA) has been overlooked and frequently persists. We discuss the privacy risks and research opportunities associated with mtDNA, urging consideration by the scientific, ethics, and legal communities.
Collapse
Affiliation(s)
- Mohamed S Sarhan
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy; Department CIBIO, University of Trento, 38123 Trento, Italy
| | - Giacomo Antonello
- Institute for Biomedicine, Eurac Research, 39100 Bolzano, Italy; Department CIBIO, University of Trento, 38123 Trento, Italy
| | - Hansi Weissensteiner
- Institute of Genetic Epidemiology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | | | | | - Levi Waldron
- Department CIBIO, University of Trento, 38123 Trento, Italy; City University of New York School of Public Health, New York, NY, USA
| | - Nicola Segata
- Department CIBIO, University of Trento, 38123 Trento, Italy.
| | | |
Collapse
|
6
|
Gridina M, Lagunov T, Belokopytova P, Torgunakov N, Nuriddinov M, Nurislamov A, Nazarenko LP, Kashevarova AA, Lopatkina ME, Vasilyev S, Zuev A, Belyaeva EO, Salyukova OA, Cheremnykh AD, Sukhanova NN, Minzhenkova ME, Markova ZG, Demina NA, Stepanchuk Y, Khabarova A, Yan A, Valeev E, Koksharova G, Grigor'eva EV, Kokh N, Lukjanova T, Maximova Y, Musatova E, Shabanova E, Kechin A, Khrapov E, Boyarskih U, Ryzhkova O, Suntsova M, Matrosova A, Karoli M, Manakhov A, Filipenko M, Rogaev E, Shilova NV, Lebedev IN, Fishman V. Combining chromosome conformation capture and exome sequencing for simultaneous detection of structural and single-nucleotide variants. Genome Med 2025; 17:47. [PMID: 40336115 PMCID: PMC12060427 DOI: 10.1186/s13073-025-01471-3] [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: 05/04/2024] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND Effective molecular diagnosis of congenital diseases hinges on comprehensive genomic analysis, traditionally reliant on various methodologies specific to each variant type-whole exome or genome sequencing for single nucleotide variants (SNVs), array CGH for copy-number variants (CNVs), and microscopy for structural variants (SVs). METHODS We introduce a novel, integrative approach combining exome sequencing with chromosome conformation capture, termed Exo-C. This method enables the concurrent identification of SNVs in clinically relevant genes and SVs across the genome and allows analysis of heterozygous and mosaic carriers. Enhanced with targeted long-read sequencing, Exo-C evolves into a cost-efficient solution capable of resolving complex SVs at base-pair accuracy. RESULTS Applied to 66 human samples Exo-C achieved 100% recall and 73% precision in detecting chromosomal translocations and SNVs. We further benchmarked its performance for inversions and CNVs and demonstrated its utility in detecting mosaic SVs and resolving diagnostically challenging cases. CONCLUSIONS Through several case studies, we demonstrate how Exo-C's multifaceted application can effectively uncover diverse causative variants and elucidate disease mechanisms in patients with rare disorders.
Collapse
Affiliation(s)
- Maria Gridina
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia.
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia.
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia.
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia.
| | - Timofey Lagunov
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Polina Belokopytova
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Nikita Torgunakov
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Miroslav Nuriddinov
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Artem Nurislamov
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
| | - Lyudmila P Nazarenko
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Anna A Kashevarova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Maria E Lopatkina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Stanislav Vasilyev
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Andrey Zuev
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Elena O Belyaeva
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Olga A Salyukova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Aleksandr D Cheremnykh
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Natalia N Sukhanova
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | | | | | - Nina A Demina
- Research Centre for Medical Genetics, Moscow, 115522, Russia
| | - Yana Stepanchuk
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Anna Khabarova
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
| | - Alexandra Yan
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Emil Valeev
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
| | - Galina Koksharova
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
| | - Elena V Grigor'eva
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
| | - Natalia Kokh
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Tatiana Lukjanova
- Center for Family Care and Reproduction, 1 Kiyevskaya Str, Novosibirsk, 6300136, Russia
| | - Yulia Maximova
- Center for Family Care and Reproduction, 1 Kiyevskaya Str, Novosibirsk, 6300136, Russia
- Novosibirsk State Medical University, Novosibirsk, 630091, Russia
| | - Elizaveta Musatova
- Genetics and Reproductive Medicine Center, "GENETICO" PJSC, Moscow, 119333, Russia
| | - Elena Shabanova
- North-Western State Medical University named after I.I. Mechnikov, Saint-Petersburg, 191015, Russia
| | - Andrey Kechin
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Evgeniy Khrapov
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Uliana Boyarskih
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Oxana Ryzhkova
- Research Centre for Medical Genetics, Moscow, 115522, Russia
| | - Maria Suntsova
- Sechenov First Moscow State Medical University, Moscow, 119435, Russia
- Endocrinology Research Center, Moscow, 117292, Russia
| | - Alina Matrosova
- Sechenov First Moscow State Medical University, Moscow, 119435, Russia
- Endocrinology Research Center, Moscow, 117292, Russia
| | - Mikhail Karoli
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
| | - Andrey Manakhov
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
| | - Maxim Filipenko
- Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, 630090, Russia
| | - Evgeny Rogaev
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia
- UMass Chan Medical School, Worcester, 01655, USA
| | | | - Igor N Lebedev
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia
| | - Veniamin Fishman
- Institute of Cytology and Genetics, 10, Prospekt Akademika Lavrent'yeva, Novosibirsk, 630090, Russia.
- Novosibirsk State University, 1, Pirogova Str, Novosibirsk, 630090, Russia.
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 10, Nab. Ushaiki, Tomsk, 634050, Russia.
- Artificial Intelligence Research Institute, Moscow, Russia, 121170.
- Sirius University of Science and Technology, Sirius Federal Territory, Sochi, 354340, Russia.
| |
Collapse
|
7
|
Li N, Gao G, Zhang T, Zhao C, Zhao Y, Zhang Y, Sun Z. Co-variation of Host Gene Expression and Gut Microbiome in Intestine-Specific Spp1 Conditional Knockout Mice. Curr Microbiol 2025; 82:282. [PMID: 40327160 PMCID: PMC12055922 DOI: 10.1007/s00284-025-04246-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/14/2025] [Indexed: 05/07/2025]
Abstract
Osteopontin, which is a highly phosphorylated and glycosylated acidic secreted protein encoded by the secreted phosphoprotein 1 (Spp1) gene, plays a crucial role in immune regulation, inflammatory responses, and cell adhesion. However, its impact on intestinal gene expression and gut microbiota remains underexplored. In this study, we developed an Spp1 conditional knockout mouse model to investigate alterations in the intestinal transcriptome and microbiome, with particular emphasis on changes in gene expression and predicted metabolic pathways. Our findings demonstrated that Spp1 gene conditional knockout significantly modified the expression of genes involved in immune regulation and lipid metabolism. Moreover, metagenomic analysis revealed marked shifts in gut microbial diversity and predicted the metabolic pathways associated with digestion, absorption, and lipid metabolism. These results suggest that Spp1 is instrumental in maintaining gut microbial equilibrium and in regulating host lipid metabolism and immune responses. This study offers new insights into the role of Spp1 in host-microbiota interactions and the potential foundations for developing related therapeutic strategies.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, People's Republic of China.
| | - Guangqi Gao
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, People's Republic of China.
| | - Tao Zhang
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, People's Republic of China
| | - Chunyan Zhao
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, People's Republic of China
| | - Yue Zhao
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, People's Republic of China
| | - Yong Zhang
- The Fourth Medical Center of PLA General Hospital, No. 51 Fucheng Road, Haidian District, Beijing, 100037, China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering (IMAU), Ministry of Education, Inner Mongolia Agricultural University, Hohhot, People's Republic of China
| |
Collapse
|
8
|
Holleman AM, Deaton AM, Hoffing RA, Krohn L, LoGerfo P, Nioi P, Plekan ME, Akle Serrano S, Ticau S, Walshe TE, Borodovsky A, Ward LD. Rare predicted loss-of-function and damaging missense variants in CFHR5 associate with protection from age-related macular degeneration. Am J Hum Genet 2025; 112:1062-1080. [PMID: 40250423 PMCID: PMC12120177 DOI: 10.1016/j.ajhg.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of blindness among older adults worldwide, but treatment options are limited. Genetics studies have implicated the CFH locus, containing CFH and five CFHR genes, CFHR1-5, in AMD. While CFH has been robustly linked with AMD risk, potential additional roles for the CFHR genes remain unclear, obscured by strong linkage disequilibrium across the locus. Investigating rare coding variants can help to identify causal genes in such regions. We used whole-exome sequencing data from 406,952 UK Biobank participants to examine AMD associations with genes at the CFH locus. For each gene, we used burden testing to examine associations of rare (minor-allele frequency [MAF] < 1%) predicted loss-of-function (pLoF) and predicted damaging missense variants with AMD. We considered "broadly defined AMD" (ICD-10 35.3; ncases = 10,700) and "strictly defined AMD" (dry or wet AMD; ncases = 346). Adjusting for CFH-region variants known to independently associate with AMD, we find that CFHR5 rare variant burden significantly associates with a decreased risk of broadly defined AMD (odds ratio [OR] = 0.75, p = 7 × 10-4), with this association primarily driven by pLoF variants. Furthermore, the association of CFHR5 rare variants with AMD protection is estimated to be stronger for individuals with the CFH rs1061170 AMD risk allele (p.Tyr402His [p.Y402H]; interaction p = 0.04). Corresponding analyses of strict AMD were underpowered. However, we observe that thinning of the photoreceptor layer outer segment strongly predicts strict AMD and find that CFHR5 rare variant burden is significantly associated with increased thickness of this retinal layer (+0.34 SD, p = 4 × 10-4, n = 45,365). These findings suggest CFHR5 inhibition as a potential therapeutic approach for AMD.
Collapse
Affiliation(s)
| | | | | | - Lynne Krohn
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | - Paul Nioi
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | | | - Simina Ticau
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| | | | | | - Lucas D Ward
- Alnylam Pharmaceuticals, Cambridge, MA 02142, USA
| |
Collapse
|
9
|
Xu L, Kun E, Pandey D, Wang JY, Brasil MF, Singh T, Narasimhan VM. The genetic architecture of and evolutionary constraints on the human pelvic form. Science 2025; 388:eadq1521. [PMID: 40208988 DOI: 10.1126/science.adq1521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 01/09/2025] [Indexed: 04/12/2025]
Abstract
Human pelvic evolution following the human-chimpanzee divergence is thought to result in an obstetrical dilemma, a mismatch between large infant brains and narrowed female birth canals, but empirical evidence has been equivocal. By using deep learning on 31,115 dual-energy x-ray absorptiometry scans from UK Biobank, we identified 180 loci associated with seven highly heritable pelvic phenotypes. Birth canal phenotypes showed sex-specific genetic architecture, aligning with reproductive function. Larger birth canals were linked to slower walking pace and reduced back pain but increased hip osteoarthritis risk, whereas narrower birth canals were associated with reduced pelvic floor disorder risk but increased obstructed labor risk. Lastly, genetic correlation between birth canal and head widths provides evidence of coevolution between the human pelvis and brain, partially mitigating the dilemma.
Collapse
Affiliation(s)
- Liaoyi Xu
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Eucharist Kun
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Devansh Pandey
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Joyce Y Wang
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
| | - Marianne F Brasil
- Department of Anthropology, Western Washington University, Bellingham, WA, USA
| | - Tarjinder Singh
- The Department of Psychiatry at Columbia University Irving Medical Center, New York, NY, USA
- The New York Genome Center, New York, NY, USA
- Mortimer B. Zuckerman Mind Brain Behavior Institute at Columbia University, New York, NY, USA
| | - Vagheesh M Narasimhan
- Department of Integrative Biology, The University of Texas at Austin, Austin, TX, USA
- Department of Statistics and Data Science, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
10
|
Claus I, Sivalingam S, Koller AC, Weiß A, Mathey CM, Sindermann L, Klein D, Henschel L, Ludwig KU, Hoffmann P, Heimbach A, Heilmann-Heimbach S, Vedder H, Kammerer-Ciernioch J, Stürmer T, Streit F, Maaser-Hecker A, Nenadić I, Baune BT, Hartmann AM, Konte B, Giegling I, Heilbronner U, Wagner M, Philipsen A, Schmidt B, Rujescu D, Buness A, Schulze TG, Rietschel M, Forstner AJ, Nöthen MM, Degenhardt F. Contribution of Rare and Potentially Functionally Relevant Sequence Variants in Schizophrenia Risk-Locus Xq28,distal. Am J Med Genet B Neuropsychiatr Genet 2025; 198:e33011. [PMID: 39473393 DOI: 10.1002/ajmg.b.33011] [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: 04/17/2024] [Revised: 09/01/2024] [Accepted: 09/17/2024] [Indexed: 03/04/2025]
Abstract
Duplications of the Xq28,distal locus have been described in male and female patients with schizophrenia (SCZ) or intellectual disability. The Xq28,distal locus spans eight protein-coding genes (F8, CMC4, MTCP1, BRCC3, VBP1, FUNDC2, CLIC2, and RAB39B) and is flanked by recurrent genomic breakpoints. Thus, the issue of which gene/s at this locus is/are relevant in terms of SCZ pathogenesis remains unclear. The aim of this study was to investigate the contribution of rare and potentially functionally relevant sequence variants within the Xq28,distal locus to SCZ risk using the single-molecule molecular inversion probes (smMIP) method. Targeted sequencing was performed in a cohort of 1935 patients with SCZ and 1905 controls of European ancestry. The consecutive statistical analysis addressed two main areas. On the level of the individual variants, allele counts in the patient and control cohort were systematically compared with a Fisher's exact test: (i) for the entire present study cohort; (ii) for patients and controls separated by sex; and (iii) in combination with data published by the Schizophrenia Exome Meta-Analysis (SCHEMA) consortium. On the gene-wise level, a burden analysis was performed using the X-chromosomal model of the Optimal Unified Sequence Kernel Association Test (SKAT-O), with adjustment for possible sex-specific effects. Targeted sequencing identified a total of 13 rare and potentially functional variants in four patients and 11 controls. However, neither at the level of individual rare and potentially functional variants nor at the level of the eight protein-coding genes at the Xq28,distal locus was a statistically significant enrichment in patients compared to controls observed. Although inconclusive, the present findings represent a step toward improved understanding of the contribution of X-chromosomal risk factors in neuropsychiatric disorder development, which is an underrepresented aspect of genetic studies in this field.
Collapse
Grants
- BONFOR Research Funding Program of the Faculty of Medicine, University of Bonn
- Dr. Lisa Oehler Foundation
- 01ZX1614K Federal Ministry of Education and Research; projects: IntegraMent and BipoLife
- 01EE1404H Federal Ministry of Education and Research; projects: IntegraMent and BipoLife
- 945151 European Union's Horizon 2020 Research and Innovation Programme; projects: PSY-PGx, GEPI-BIOPSY and MulioBio
- 01EW2005 European Union's Horizon 2020 Research and Innovation Programme; projects: PSY-PGx, GEPI-BIOPSY and MulioBio
- 01EW2009 European Union's Horizon 2020 Research and Innovation Programme; projects: PSY-PGx, GEPI-BIOPSY and MulioBio
- 514201724 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
- STU 235/10-2 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
- HE 2443/8-1 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
- AM37/19-1 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
- SCHU1603/4-1,5-1,7-1 German Research Foundation; Projects: PsyCourse, KFO 241 and Heidelberg Cohort Study of the Elderly
Collapse
Affiliation(s)
- I Claus
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - S Sivalingam
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
- Core Unit for Bioinformatics Data Analysis, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Human Genetics, Medical Faculty, University Hospital of Düsseldorf, Heinrich Heine University of Düsseldorf, Düsseldorf, Germany
| | - A C Koller
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - A Weiß
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - C M Mathey
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - L Sindermann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - D Klein
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - L Henschel
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - K U Ludwig
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - P Hoffmann
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Genomics, Life & Brain Center, University of Bonn, Bonn, Germany
- Division of Medical Genetics, University Hospital and Department of Biomedicine, University of Basel, Basel, Switzerland
| | - A Heimbach
- NGS Core Facility, Medical Faculty, University of Bonn, Bonn, Germany
| | - S Heilmann-Heimbach
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - H Vedder
- Psychiatric Center Nordbaden, Wiesloch, Germany
| | | | - T Stürmer
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, USA
| | - F Streit
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - A Maaser-Hecker
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Genetics and Aging Research Unit, Department of Neurology, Mass General Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- McCance Center for Brain Health, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - I Nenadić
- Department of Psychiatry und Psychotherapy, University of Marburg, Marburg, Germany
- Center for Mind, Brain and Behavior (CMBB), University of Marburg, Marburg, Germany
- Department of Psychiatry and Psychotherapy, Jena University Hospital, Jena, Germany
| | - B T Baune
- Department of Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Central Melbourne, Australia
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Central Melbourne, Australia
| | - A M Hartmann
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - B Konte
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - I Giegling
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - U Heilbronner
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
| | - M Wagner
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - A Philipsen
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - B Schmidt
- Institute for Medical Informatics, Biometry and Epidemiology (IMIBE), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - D Rujescu
- Department of Psychiatry and Psychotherapy, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - A Buness
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University of Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
- Core Unit for Bioinformatics Data Analysis, Medical Faculty, University of Bonn, Bonn, Germany
| | - T G Schulze
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, Munich, Germany
- Department of Psychiatry and Behavioral Sciences, SUNY Upstate Medical University, Syracuse, New York, USA
| | - M Rietschel
- Department of Genetic Epidemiology in Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - A J Forstner
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Institute of Neuroscience and Medicine (INM-1), Research Center Jülich, Jülich, Germany
| | - M M Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
| | - F Degenhardt
- Institute of Human Genetics, University of Bonn, School of Medicine and University Hospital Bonn, Bonn, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
11
|
Abbasova L, Urbanaviciute P, Hu D, Ismail JN, Schilder BM, Nott A, Skene NG, Marzi SJ. CUT&Tag recovers up to half of ENCODE ChIP-seq histone acetylation peaks. Nat Commun 2025; 16:2993. [PMID: 40148272 PMCID: PMC11950320 DOI: 10.1038/s41467-025-58137-2] [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: 07/28/2022] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
DNA-protein interactions have traditionally been profiled via chromatin immunoprecipitation followed by next-generation sequencing (ChIP-seq). Cleavage Under Targets & Tagmentation (CUT&Tag) is a rapidly expanding technique that enables the profiling of such interactions in situ at high sensitivity. However, thorough evaluation and benchmarking against established ChIP-seq datasets are lacking. Here, we comprehensively benchmarked CUT&Tag for H3K27ac and H3K27me3 against published ChIP-seq profiles from ENCODE in K562 cells. Combining multiple new and published CUT&Tag datasets, there was an average recall of 54% known ENCODE peaks for both histone modifications. We tested peak callers MACS2 and SEACR and identified optimal peak calling parameters. Overall, peaks identified by CUT&Tag represent the strongest ENCODE peaks and show the same functional and biological enrichments as ChIP-seq peaks identified by ENCODE. Our workflow systematically evaluates the merits of methodological adjustments, providing a benchmarking framework for the experimental design and analysis of CUT&Tag studies.
Collapse
Affiliation(s)
- Leyla Abbasova
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Paulina Urbanaviciute
- UK Dementia Research Institute at King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Di Hu
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Joy N Ismail
- UK Dementia Research Institute at King's College London, London, UK
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Brian M Schilder
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Alexi Nott
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Nathan G Skene
- UK Dementia Research Institute at Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Sarah J Marzi
- Department of Brain Sciences, Imperial College London, London, UK.
- UK Dementia Research Institute at King's College London, London, UK.
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
| |
Collapse
|
12
|
Satas G, Myers MA, McPherson A, Shah SP. Inferring active mutational processes in cancer using single cell sequencing and evolutionary constraints. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639589. [PMID: 40060559 PMCID: PMC11888314 DOI: 10.1101/2025.02.24.639589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
Ongoing mutagenesis in cancer drives genetic diversity throughout the natural history of cancers. As the activities of mutational processes are dynamic throughout evolution, distinguishing the mutational signatures of 'active' and 'historical' processes has important implications for studying how tumors evolve. This can aid in understanding mutagenic states at the time of presentation, and in associating active mutational process with therapeutic resistance. As bulk sequencing primarily captures historical mutational processes, we studied whether ultra-low-coverage single-cell whole-genome sequencing (scWGS), which measures the distribution of mutations across hundreds or thousands of individual cells, could enable the distinction between historical and active mutational processes. While technical challenges and data sparsity have limited mutation analysis in scWGS, we show that these data contain valuable information about dynamic mutational processes. To robustly interpret single nucleotide variants (SNVs) in scWGS, we introduce ArtiCull, a method to identify and remove SNV artifacts by leveraging evolutionary constraints, enabling reliable detection of mutations for signature analysis. Applying this approach to scWGS data from pancreatic ductal adenocarcinoma (PDAC), triple-negative breast cancer (TNBC), and high-grade serous ovarian cancer (HGSOC), we uncover temporal and spatial patterns in mutational processes. In PDAC, we observe a temporal increase in mismatch repair deficiency (MMRd). In cisplatin-treated TNBC patient-derived xenografts, we identify therapy-induced mutagenesis and inactivation of APOBEC3 activity. In HGSOC, we show distinct patterns of APOBEC3 mutagenesis, including late tumor-wide activation in one case and clade-specific enrichment in another. Additionally, we detect a clone-specific increase in SBS17 activity, in a clone previously linked to recurrence. Our findings establish ultra-low-coverage scWGS as a powerful approach for studying active mutational processes that may influence ongoing clonal evolution and therapeutic resistance.
Collapse
Affiliation(s)
- Gryte Satas
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew A. Myers
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew McPherson
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sohrab P. Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
13
|
Marsh WA, Hall A, Barnes I, Price B. Facilitating high throughput collections-based genomics: a comparison of DNA extraction and library building methods. Sci Rep 2025; 15:6013. [PMID: 39972011 PMCID: PMC11839992 DOI: 10.1038/s41598-025-88443-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/28/2025] [Indexed: 02/21/2025] Open
Abstract
While DNA barcoding methods are an increasingly important tool in biological conservation, the resource requirements of constructing reference libraries frequently reduce their efficacy. One efficient way of sourcing taxonomically validated DNA for reference libraries is to use museum collections. However, DNA degradation intrinsic to historical museum specimens can, if not addressed in the wet lab, lead to low quality data generation and severely limit scientific output. Several DNA extraction and library build methods that are designed to work with degraded DNA have been developed, although the ability to implement these methods at scale and at low cost has yet to be formally addressed. Here, the performance of widely used DNA extraction and library build methods are compared using museum specimens. We find that while our selected DNA extraction methods do not significantly differ in DNA yield, the Santa Cruz Reaction (SCR) library build method is not only the most effective at retrieving degraded DNA from museum specimens but also easily implemented at high throughput for low cost. Results highlight the importance of lab protocol on data yield. An optimised "sample to sequencing" high-throughput protocol which incorporates SCR is included to allow for easy uptake by the wider scientific community.
Collapse
Affiliation(s)
- William A Marsh
- Natural History Museum, Cromwell Road, South Kensington, SW7 5BD, London, UK.
| | - Andie Hall
- Natural History Museum, Cromwell Road, South Kensington, SW7 5BD, London, UK
| | - Ian Barnes
- Natural History Museum, Cromwell Road, South Kensington, SW7 5BD, London, UK
| | - Ben Price
- Natural History Museum, Cromwell Road, South Kensington, SW7 5BD, London, UK
| |
Collapse
|
14
|
Chao KH, Heinz JM, Hoh C, Mao A, Shumate A, Pertea M, Salzberg SL. Combining DNA and protein alignments to improve genome annotation with LiftOn. Genome Res 2025; 35:311-325. [PMID: 39730188 PMCID: PMC11874971 DOI: 10.1101/gr.279620.124] [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: 05/24/2024] [Accepted: 12/19/2024] [Indexed: 12/29/2024]
Abstract
As the number and variety of assembled genomes continue to grow, the number of annotated genomes is falling behind, particularly for eukaryotes. DNA-based mapping tools help to address this challenge, but they are only able to transfer annotation between closely related species. Here we introduce LiftOn, a homology-based software tool that integrates DNA and protein alignments to enhance the accuracy of genome-scale annotation and to allow mapping between relatively distant species. LiftOn's protein-centric algorithm considers both types of alignments, chooses optimal open reading frames, resolves overlapping gene loci, and finds additional gene copies when they exist. LiftOn can reliably transfer annotation between genomes representing members of the same species, as we demonstrate on human, mouse, honeybee, rice, and Arabidopsis thaliana It can further map annotation effectively across species pairs as far apart as mouse and rat or Drosophila melanogaster and Drosophila erecta.
Collapse
Affiliation(s)
- Kuan-Hao Chao
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA;
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Jakob M Heinz
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Celine Hoh
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Alan Mao
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Alaina Shumate
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Mihaela Pertea
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Steven L Salzberg
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA;
- Center for Computational Biology, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
- Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland 21211, USA
| |
Collapse
|
15
|
Shallangwa SM, Ross AW, Morgan PJ. Single, but not mixed dietary fibers suppress body weight gain and adiposity in high fat-fed mice. Front Microbiol 2025; 16:1544433. [PMID: 40012787 PMCID: PMC11861375 DOI: 10.3389/fmicb.2025.1544433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 01/13/2025] [Indexed: 02/28/2025] Open
Abstract
Dietary fiber can suppress excess adipose tissue and weight gain in rodents and humans when fed high fat diets. The gut microbiome is thought to have a key role, although exactly how remains unclear. In a tightly controlled murine study, we explored how different types of dietary fiber and doses affect the gut microbiota and gut epithelial gene expression. We show that 10% pectin and 10% FOS suppress high fat diet (HFD)-induced weight gain, effects not seen at 2% doses. Furthermore, 2 and 10% mixtures of dietary fiber were also without effect. Each fiber treatment stimulated a distinct gut microbiota profile at the family and operational taxonomic unit (OTU) level. Mechanistically it is likely that the single 10% fiber dose shifted selected bacteria above some threshold abundance, required to suppress body weight, which was not achieved by the 10% Mix, composed of 4 fibers each at 2.5%. Plasma levels of the gut hormone PYY were elevated by 10% pectin and FOS, but not 10% mixed fibers, and similarly RNA seq revealed some distinct effects of the 10% single fibers on gut epithelial gene expression. These data show how the ability of dietary fiber to suppress HFD-induced weight gain is dependent upon both fiber type and dose. It also shows that the microbial response to dietary fiber is distinct and that there is not a single microbial response associated with the inhibition of adiposity and weight gain. PYY seems key to the latter response, although the role of other factors such as Reg3γ and CCK needs to be explored.
Collapse
Affiliation(s)
| | | | - Peter J. Morgan
- Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
16
|
Zanghí G, Patel H, Smith JL, Camargo N, Bae Y, Hesping E, Boddey JA, Venugopal K, Marti M, Flannery EL, Chuenchob V, Fishbaugher ME, Mikolajczak SA, Roobsoong W, Sattabongkot J, Gupta P, Pazzagli L, Rezakhani N, Betz W, Hayes K, Goswami D, Vaughan AM, Kappe SHI. Genome-wide gene expression profiles throughout human malaria parasite liver stage development in humanized mice. Nat Microbiol 2025; 10:569-584. [PMID: 39891010 PMCID: PMC11790487 DOI: 10.1038/s41564-024-01905-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/04/2024] [Indexed: 02/03/2025]
Abstract
Gene expression of Plasmodium falciparum (Pf) liver-stage (LS) parasites has remained poorly characterized, although they are major vaccine and drug targets. Using a human liver-chimaeric mouse model and a fluorescent parasite line (PfNF54CSPGFP), we isolated PfLS and performed transcriptomics on key LS developmental phases. We linked clustered gene expression to ApiAP2, a major family of transcription factors that regulate the parasite life cycle. This provided insights into transcriptional regulation of LS infection and expression of essential LS metabolic and biosynthetic pathways. We observed expression of antigenically variant PfEMP1 proteins and the major Pf protein export machine PTEX and identified protein candidates that might be exported by LS parasites. Comparing Pf and P. vivax LS transcriptomes, we uncovered differences in their expression of sexual commitment factors. This data will aid LS research and vaccine and drug target identification for prevention of malaria infection.
Collapse
Affiliation(s)
- Gigliola Zanghí
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
| | - Hardik Patel
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Jenny L Smith
- Research Scientific Computing, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nelly Camargo
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Yeji Bae
- Research Scientific Computing, Seattle Children's Research Institute, Seattle, WA, USA
| | - Eva Hesping
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Justin A Boddey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kannan Venugopal
- Institute for Parasitology, University of Zurich, Zurich, Switzerland
- Institute of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Matthias Marti
- Institute for Parasitology, University of Zurich, Zurich, Switzerland
- Institute of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Erika L Flannery
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Vorada Chuenchob
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Matthew E Fishbaugher
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
- Global Health, Biomedical Research, Novartis, Emeryville, CA, USA
| | - Sebastian A Mikolajczak
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Priya Gupta
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Lucia Pazzagli
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Nastaran Rezakhani
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - William Betz
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Kiera Hayes
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Debashree Goswami
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Ashley M Vaughan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Stefan H I Kappe
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
17
|
Abdulai-Saiku S, Gupta S, Wang D, Marino F, Moreno AJ, Huang Y, Srivastava D, Panning B, Dubal DB. The maternal X chromosome affects cognition and brain ageing in female mice. Nature 2025; 638:152-159. [PMID: 39843739 PMCID: PMC11798838 DOI: 10.1038/s41586-024-08457-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 11/27/2024] [Indexed: 01/24/2025]
Abstract
Female mammalian cells have two X chromosomes, one of maternal origin and one of paternal origin. During development, one X chromosome randomly becomes inactivated1-4. This renders either the maternal X (Xm) chromosome or the paternal X (Xp) chromosome inactive, causing X mosaicism that varies between female individuals, with some showing considerable or complete skew of the X chromosome that remains active5-7. Parent-of-X origin can modify epigenetics through DNA methylation8,9 and possibly gene expression; thus, mosaicism could buffer dysregulated processes in ageing and disease. However, whether X skew or its mosaicism alters functions in female individuals is largely unknown. Here we tested whether skew towards an active Xm chromosome influences the brain and body-and then delineated unique features of Xm neurons and Xp neurons. An active Xm chromosome impaired cognition in female mice throughout the lifespan and led to worsened cognition with age. Cognitive deficits were accompanied by Xm-mediated acceleration of biological or epigenetic ageing of the hippocampus, a key centre for learning and memory, in female mice. Several genes were imprinted on the Xm chromosome of hippocampal neurons, suggesting silenced cognitive loci. CRISPR-mediated activation of Xm-imprinted genes improved cognition in ageing female mice. Thus, the Xm chromosome impaired cognition, accelerated brain ageing and silenced genes that contribute to cognition in ageing. Understanding how Xm impairs brain function could lead to an improved understanding of heterogeneity in cognitive health in female individuals and to X-chromosome-derived pathways that protect against cognitive deficits and brain ageing.
Collapse
Affiliation(s)
- Samira Abdulai-Saiku
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Shweta Gupta
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Dan Wang
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Francesca Marino
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Neurosciences Graduate Program, University of California San Francisco, San Francisco, CA, USA
| | - Arturo J Moreno
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Yu Huang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Barbara Panning
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Dena B Dubal
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
- Neurosciences Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- Bakar Aging Research Institute, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
18
|
Alagöz G, Eising E, Mekki Y, Bignardi G, Fontanillas P, Nivard MG, Luciano M, Cox NJ, Fisher SE, Gordon RL. The shared genetic architecture and evolution of human language and musical rhythm. Nat Hum Behav 2025; 9:376-390. [PMID: 39572686 PMCID: PMC11860242 DOI: 10.1038/s41562-024-02051-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: 10/02/2023] [Accepted: 10/07/2024] [Indexed: 11/27/2024]
Abstract
This study aimed to test theoretical predictions over biological underpinnings of previously documented phenotypic correlations between human language-related and musical rhythm traits. Here, after identifying significant genetic correlations between rhythm, dyslexia and various language-related traits, we adapted multivariate methods to capture genetic signals common to genome-wide association studies of rhythm (N = 606,825) and dyslexia (N = 1,138,870). The results revealed 16 pleiotropic loci (P < 5 × 10-8) jointly associated with rhythm impairment and dyslexia, and intricate shared genetic and neurobiological architectures. The joint genetic signal was enriched for foetal and adult brain cell-specific regulatory regions, highlighting complex cellular composition in their shared underpinnings. Local genetic correlation with a key white matter tract (the left superior longitudinal fasciculus-I) substantiated hypotheses about auditory-motor connectivity as a genetically influenced, evolutionarily relevant neural endophenotype common to rhythm and language processing. Overall, we provide empirical evidence of multiple aspects of shared biology linking language and musical rhythm, contributing novel insight into the evolutionary relationships between human musicality and linguistic communication traits.
Collapse
Affiliation(s)
- Gökberk Alagöz
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands.
| | - Else Eising
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
| | - Yasmina Mekki
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Giacomo Bignardi
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands
- Max Planck School of Cognition, Leipzig, Germany
| | | | - Michel G Nivard
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Michelle Luciano
- Department of Psychology, University of Edinburgh, Edinburgh, UK
| | - Nancy J Cox
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Nijmegen, the Netherlands.
- Donders Institute for Brain, Cognition and Behaviour, Radboud University, Nijmegen, the Netherlands.
| | - Reyna L Gordon
- Department of Otolaryngology-Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA.
- Department of Hearing & Speech Sciences, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
19
|
Prideaux EB, Boyle DL, Choi E, Buckner JH, Robinson WH, Holers VM, Deane KD, Firestein GS, Wang W. Epigenetic trajectory predicts development of clinical rheumatoid arthritis in ACPA+ individuals: Targeting Immune Responses for Prevention of Rheumatoid Arthritis (TIP-RA). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.15.618490. [PMID: 39463978 PMCID: PMC11507690 DOI: 10.1101/2024.10.15.618490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
OBJECTIVE The presence of autoantibodies to citrullinated protein antigens (ACPAs) in the absence of clinically-apparent inflammatory arthritis (IA) identifies individuals at-risk for developing future clinical rheumatoid arthritis (RA). However, it is unclear why some ACPA+ individuals convert to clinical RA while others do not. We explored the possibility in the Targeting Immune Responses for Prevention of Rheumatoid Arthritis (TIP-RA) study that epigenetic remodeling is part of the trajectory from an at-risk state to clinical disease and identifies novel biomarkers associated with conversion to clinical RA. METHODS ACPA- Controls, ACPA+ At-Risk, and Early RA individuals were followed for up to 5 years, including obtaining blood samples annually and at RA diagnosis. Peripheral blood mononuclear cells (PBMCs) were separated into CD19+ B cells, memory CD4+ T cells, and naive CD4+ T cells using antibodies and magnetic beads. Genome-wide methylation within each cell lineage was assayed using the Illumina MethylationEPIC v1.0 beadchip. ACPA+ At-Risk participants who did or did not develop RA were designated Pre-RA or Non-converters, respectively. Differentially methylated loci (DML) were selected using the Limma software package. Using the Caret package, we constructed machine learning models in test and validation cohorts and identified the most predictive loci of clinical RA conversion. RESULTS Cross-sectional differential methylation analysis at baseline revealed DMLs that distinguish the Pre-RA methylome from ACPA+ Non-converters, the latter which closely resembled ACPA- Controls. Genes overlapping these DMLs correspond to aberrant NOTCH signaling and DNA repair pathways in B cells. Longitudinal analysis showed that ACPA- Control and ACPA+ Non-converter methylomes are relatively constant. In contrast, the Pre-RA methylome remodeled along a dynamic RA methylome trajectory characterized by epigenetic changes in active regulatory elements. Clinical conversion to RA, defined based on diagnosis, marked an epigenetic inflection point for cell cycle pathways in B cells and adaptive immunity pathways in naive T cells. Machine learning revealed individual loci associated with RA conversion. This model significantly outperformed autoantibodies plus acute phase reactants as predictors of RA conversion. CONCLUSION DNA methylation is a dynamic process in ACPA+ individuals at-risk for developing RA that eventually transition to clinical disease. In contrast, non-converters and controls have stable methylomes. The accumulation of epigenetic marks over time prior to conversion to clinical RA conforms to pathways that are associated with immunity and can be used to identify potential pathogenic pathways for therapeutic targeting and/or use as prognostic biomarkers.
Collapse
|
20
|
Mohseni Ahooyi T, Stear B, Simmons JA, Nemarich CM, Silverstein JC, Taylor DM. Homo Sapiens Chromosomal Location Ontology: A Framework for Genomic Data in Biomedical Knowledge Graphs. Sci Data 2025; 12:52. [PMID: 39799122 PMCID: PMC11724927 DOI: 10.1038/s41597-024-04358-x] [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: 02/21/2024] [Accepted: 12/20/2024] [Indexed: 01/15/2025] Open
Abstract
The Homo sapiens Chromosomal Location Ontology (HSCLO) is designed to facilitate the integration of human genomic features into biomedical knowledge graphs from releases GRCh37 and GRCh38 at multiple resolutions. HSCLO comprises two distinct versions, HSCLO37 and HSCLO38, each tailored to its respective human genome release. This ontology supports the efficient integration and analysis of human genomic data across scales ranging from entire chromosomes to individual base pairs, thereby enhancing data retrieval and interoperability within large-scale biomedical datasets. Unlike existing ontologies that primarily focus on genomic feature identification or annotation, HSCLO is specifically engineered to optimize the interoperability and scalability of genomic data within biomedical knowledge graphs. The utility and performance of HSCLO are demonstrated through a case study involving the integration of high-resolution chromatin interaction data, which reveals significant improvements in query efficiency and data linkage. HSCLO represents a valuable resource for advancing research in disease genetics, personalized medicine, and other domains that require complex genomic data integration.
Collapse
Affiliation(s)
- Taha Mohseni Ahooyi
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Benjamin Stear
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - J Alan Simmons
- Department of Biomedical Informatics, School of Medicine, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Christopher M Nemarich
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jonathan C Silverstein
- Department of Biomedical Informatics, School of Medicine, The University of Pittsburgh, Pittsburgh, PA, USA
| | - Deanne M Taylor
- The Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, University of Pennsylvania Perelman Medical School, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Kim J, Park J, Yang J, Kim S, Joe S, Park G, Hwang T, Cho MJ, Lee S, Lee JE, Park JH, Yeo MK, Kim SY. Highly accurate Korean draft genomes reveal structural variation highlighting human telomere evolution. Nucleic Acids Res 2025; 53:gkae1294. [PMID: 39778865 PMCID: PMC11707537 DOI: 10.1093/nar/gkae1294] [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: 05/07/2023] [Revised: 12/09/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
Given the presence of highly repetitive genomic regions such as subtelomeric regions, understanding human genomic evolution remains challenging. Recently, long-read sequencing technology has facilitated the identification of complex genetic variants, including structural variants (SVs), at the single-nucleotide level. Here, we resolved SVs and their underlying DNA damage-repair mechanisms in subtelomeric regions, which are among the most uncharted genomic regions. We generated ∼20 × high-fidelity long-read sequencing data from three Korean individuals and their partially phased high-quality de novo genome assemblies (contig N50: 6.3-58.2 Mb). We identified 131 138 deletion and 121 461 insertion SVs, 41.6% of which were prevalent in the East Asian population. The commonality of the SVs identified among the Korean population was examined by short-read sequencing data from 103 Korean individuals, providing the first comprehensive SV set representing the population based on the long-read assemblies. Manual investigation of 19 large subtelomeric SVs (≥5 kb) and their associated repair signatures revealed the potential repair mechanisms leading to the formation of these SVs. Our study provides mechanistic insight into human telomere evolution and can facilitate our understanding of human SV formation.
Collapse
Affiliation(s)
- Jun Kim
- Department of Convergent Bioscience and Informatics, College of Bioscience and Biotechnology, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jong Lyul Park
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience & Biotechnology, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Bioscience, University of Science and Technology (UST), 217, Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Jin Ok Yang
- Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science & Technology (KAIST), 291, Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Sangok Kim
- Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Bioscience, University of Science and Technology (UST), 217, Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Soobok Joe
- Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Gunwoo Park
- Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Taeyeon Hwang
- Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Mun-Jeong Cho
- Department of Bioscience, University of Science and Technology (UST), 217, Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Seungjae Lee
- DNALink, Inc, 31, Magokjungang 8-ro 3-gil, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Jong-Eun Lee
- DNALink, Inc, 31, Magokjungang 8-ro 3-gil, Gangseo-gu, Seoul 07793, Republic of Korea
| | - Ji-Hwan Park
- Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Biological Science, Ajou University, 206, World cup-ro, Yeongtong-gu, Suwon 16499, Republic of Korea
| | - Min-Kyung Yeo
- Department of Pathology, Chungnam National University School of Medicine, 282, Munhwa-ro, Jung-gu, Daejeon 35015, Republic of Korea
| | - Seon-Young Kim
- Korea Bioinformation Center, Korea Research Institute of Bioscience & Biotechnology, 125, Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Bioscience, University of Science and Technology (UST), 217, Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| |
Collapse
|
22
|
Shi W, Jin E, Fang L, Sun Y, Fan Z, Zhu J, Liang C, Zhang YP, Zhang YQ, Wang GD, Zhao W. VDGE: a data repository of variation database for gene-edited animals across multiple species. Nucleic Acids Res 2025; 53:D1250-D1260. [PMID: 39470732 PMCID: PMC11701559 DOI: 10.1093/nar/gkae956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024] Open
Abstract
Gene-edited animals are crucial for addressing fundamental questions in biology and medicine and hold promise for practical applications. In light of the rapid advancement of gene editing technologies over the past decade, a dramatically increased number of gene-edited animals have been generated. Genome editing at off-target sites can, however, introduce genomic variations, potentially leading to unintended functional consequences in these animals. So, there is an urgent need to systematically collect and collate these variations in gene-edited animals to aid data mining and integrative in-depth analyses. However, existing databases are currently insufficient to meet this need. Here, we present the Variation Database of Gene-Edited animals (VDGE, https://ngdc.cncb.ac.cn/vdge), the first open-access repository to present genomic variations and annotations in gene-edited animals, with a particular focus on larger animals such as monkeys. At present, VDGE houses 151 on-target mutations from 210 samples, and 115,710 variations identified from 107 gene-edited and wild-type animal trios through unified and standardized analysis and concurrently provides comprehensive annotation details for each variation, thus facilitating the assessment of their functional consequences and promoting mechanistic studies and practical applications for gene-edited animals.
Collapse
Affiliation(s)
- Wenwen Shi
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Enhui Jin
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| | - Lu Fang
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Yanling Sun
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Lester and Sue Smith Breast Center, Baylor College of Medicine, One Baylor Plaza, Cambridge Street, Houston, TX 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Cambridge Street, Houston, TX 77030, USA
| | - Zhuojing Fan
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Junwei Zhu
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Chengzhi Liang
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
| | - Ya-Ping Zhang
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Panlong District, Kunming 650201, China
| | - Yong Q Zhang
- State Key Laboratory of Molecular and Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- School of Life Sciences, Hubei University, 368 Youyi Avenue, Wuchang District, Wuhan 430062, China
| | - Guo-Dong Wang
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
- Key Laboratory of Genetic Evolution and Animal Models, Yunnan Key Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, No.17 Longxin Road, Panlong District, Kunming 650201, China
| | - Wenming Zhao
- National Genomics Data Center, China National Center for Bioinformation, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, No. 1 West Beichen Road, Chaoyang District, Beijing 100101, China
- University of Chinese Academy of Sciences, No.1 Yanqihu East Rd, Huairou District, Beijing 101408, China
| |
Collapse
|
23
|
Suzuki T, Ninomiya K, Funayama T, Okamura Y, Tadaka S, Kinoshita K, Yamamoto M, Kure S, Kikuchi A, Tamiya G, Takayama J. Next-generation sequencing analysis with a population-specific human reference genome. Genes Genet Syst 2024; 99:n/a. [PMID: 39462538 DOI: 10.1266/ggs.24-00112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024] Open
Abstract
Next-generation sequencing (NGS) has become widely available and is routinely used in basic research and clinical practice. The reference genome sequence is an essential resource for NGS analysis, and several population-specific reference genomes have recently been constructed to provide a choice to deal with the vast genetic diversity of human samples. However, resources supporting population-specific references are insufficient, and it is burdensome to perform analysis using these reference genomes. Here, we constructed a set of resources to support NGS analysis using the Japanese reference genome, JG. We created resources for variant calling, variant effect prediction, gene and repeat element annotations, read mappability and RNA-seq analysis. We also provide a resource for reference coordinate conversion for further annotation enrichment. We then provide a variant calling protocol with JG. Our resources provide a guide to prepare sufficient resources for the use of population-specific reference genomes and can facilitate the migration of reference genomes.
Collapse
Affiliation(s)
- Tomohisa Suzuki
- Department of AI and Innovative Medicine, Tohoku University School of Medicine
- Department of Pediatrics, Tohoku University School of Medicine
| | - Kota Ninomiya
- Department of AI and Innovative Medicine, Tohoku University School of Medicine
- Deceased July 13, 2024
| | - Takamitsu Funayama
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
- RIKEN Center for Advanced Intelligence Project
| | - Yasunobu Okamura
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University
| | - Shu Tadaka
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
| | - Kengo Kinoshita
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University
- Tohoku Medical Megabank Organization, Tohoku University
- Department of Applied Information Sciences, Graduate School of Information Sciences, Tohoku University
- Department of In Silico Analyses, Institute of Development, Aging and Cancer, Tohoku University
| | - Masayuki Yamamoto
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University
| | - Shigeo Kure
- Department of Pediatrics, Tohoku University School of Medicine
- Miyagi Children's Hospital
| | - Atsuo Kikuchi
- Department of Pediatrics, Tohoku University School of Medicine
| | - Gen Tamiya
- Department of AI and Innovative Medicine, Tohoku University School of Medicine
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
- RIKEN Center for Advanced Intelligence Project
| | - Jun Takayama
- Department of AI and Innovative Medicine, Tohoku University School of Medicine
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University
- RIKEN Center for Advanced Intelligence Project
| |
Collapse
|
24
|
van Lingen HJ, Suarez-Diez M, Saccenti E. Normalization of gene counts affects principal components-based exploratory analysis of RNA-sequencing data. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2024; 1867:195058. [PMID: 39154857 DOI: 10.1016/j.bbagrm.2024.195058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/25/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
Normalization of gene expression count data is an essential step of in the analysis of RNA-sequencing data. Its statistical analysis has been mostly addressed in the context of differential expression analysis, that is in the univariate setting. However, relationships among genes and samples are better explored and quantified using multivariate exploratory data analysis tools like Principal Component Analysis (PCA). In this study we investigate how normalization impacts the PCA model and its interpretation, considering twelve different widely used normalization methods that were applied on simulated and experimental data. Correlation patterns in the normalized data were explored using both summary statistics and Covariance Simultaneous Component Analysis. The impact of normalization on the PCA solution was assessed by exploring the model complexity, the quality of sample clustering in the low-dimensional PCA space and gene ranking in the model fit to normalized data. PCA models upon normalization were interpreted in the context gene enrichment pathway analysis. We found that although PCA score plots are often similar independently form the normalization used, biological interpretation of the models can depend heavily on the normalization method applied.
Collapse
Affiliation(s)
- Henk J van Lingen
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, the Netherlands
| | - Maria Suarez-Diez
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, the Netherlands
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, the Netherlands.
| |
Collapse
|
25
|
Wilhelm SDP, Moresco AA, Rivero AD, Siu VM, Heinemann IU. Characterization of a novel heterozygous variant in the histidyl-tRNA synthetase gene associated with Charcot-Marie-Tooth disease type 2W. IUBMB Life 2024; 76:1125-1138. [PMID: 39352000 PMCID: PMC11580374 DOI: 10.1002/iub.2918] [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: 06/19/2024] [Accepted: 09/01/2024] [Indexed: 10/03/2024]
Abstract
Heterozygous pathogenic variants in the histidyl-tRNA synthetase (HARS) gene are associated with Charcot-Marie-Tooth (CMT) type 2W disease, classified as an axonal peripheral neuropathy. To date, at least 60 variants causing CMT symptoms have been identified in seven different aminoacyl-tRNA synthetases, with eight being found in the catalytic domain of HARS. The genetic data clearly show a causative role of aminoacyl-tRNA synthetases in CMT; however, the cellular mechanisms leading to pathology can vary widely and are unknown in the case of most identified variants. Here we describe a novel HARS variant, c.412T>C; p.Y138H, identified through a CMT gene panel in a patient with peripheral neuropathy. To determine the effect of p.Y138H we employed a humanized HARS yeast model and recombinant protein biochemistry, which identified a deficiency in protein dimerization and a growth defect which shows mild but significant improvement with histidine supplementation. This raises the potential for a clinical trial of histidine.
Collapse
Affiliation(s)
- Sarah D. P. Wilhelm
- Department of BiochemistryThe University of Western OntarioLondonOntarioCanada
| | - Angelica A. Moresco
- Division of Medical Genetics, Department of PaediatricsThe University of Western OntarioLondonOntarioCanada
| | | | - Victoria Mok Siu
- Division of Medical Genetics, Department of PaediatricsThe University of Western OntarioLondonOntarioCanada
- Children's Health Research InstituteLondonOntarioCanada
| | - Ilka U. Heinemann
- Department of BiochemistryThe University of Western OntarioLondonOntarioCanada
- Children's Health Research InstituteLondonOntarioCanada
| |
Collapse
|
26
|
Mukhametzyanova L, Schmitt LT, Torres-Rivera J, Rojo-Romanos T, Lansing F, Paszkowski-Rogacz M, Hollak H, Brux M, Augsburg M, Schneider PM, Buchholz F. Activation of recombinases at specific DNA loci by zinc-finger domain insertions. Nat Biotechnol 2024; 42:1844-1854. [PMID: 38297187 PMCID: PMC11631766 DOI: 10.1038/s41587-023-02121-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: 04/18/2023] [Accepted: 12/22/2023] [Indexed: 02/02/2024]
Abstract
Recombinases have several potential advantages as genome editing tools compared to nucleases and other editing enzymes, but the process of engineering them to efficiently recombine predetermined DNA targets demands considerable investment of time and labor. Here we sought to harness zinc-finger DNA-binding domains (ZFDs) to program recombinase binding by developing fusions, in which ZFDs are inserted into recombinase coding sequences. By screening libraries of hybrid proteins, we optimized the insertion site, linker length, spacing and ZFD orientation and generated Cre-type recombinases that remain dormant unless the insertionally fused ZFD binds its target site placed in the vicinity of the recombinase binding site. The developed fusion improved targeted editing efficiencies of recombinases by four-fold and abolished measurable off-target activity in mammalian cells. The ZFD-dependent activity is transferable to a recombinase with relaxed specificity, providing the means for developing fully programmable recombinases. Our engineered recombinases provide improved genome editing tools with increased precision and efficiency.
Collapse
Affiliation(s)
- Liliya Mukhametzyanova
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Lukas Theo Schmitt
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | - Julia Torres-Rivera
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Teresa Rojo-Romanos
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | - Felix Lansing
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | | | - Heike Hollak
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | - Melanie Brux
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Martina Augsburg
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
| | - Paul Martin Schneider
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany
- Seamless Therapeutics GmbH, Dresden, Germany
| | - Frank Buchholz
- Medical Systems Biology, Medical Faculty, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
27
|
Adams M, Vollmers C. Generation and analysis of a mouse multitissue genome annotation atlas. Genome Res 2024; 34:2108-2117. [PMID: 39443154 DOI: 10.1101/gr.279217.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 09/11/2024] [Indexed: 10/25/2024]
Abstract
Generating an accurate and complete genome annotation for an organism is complex because the cells within each tissue can express a unique set of transcript isoforms from a unique set of genes. A comprehensive genome annotation should contain information on what tissues express what transcript isoforms at what level. This tissue-level isoform information can then inform a wide range of research questions as well as experiment designs. Long-read sequencing technology combined with advanced full-length cDNA library preparation methods has now achieved throughput and accuracy where generating these types of annotations is achievable. Here, we show this by generating a genome annotation of the mouse (Mus musculus). We used the nanopore-based R2C2 long-read sequencing method to generate 64 million highly accurate full-length cDNA consensus reads-averaging 5.4 million reads per tissue for a dozen tissues. Using the Mandalorion tool, we processed these reads to generate the Tissue-level Atlas of Mouse Isoforms which is available as a trackhub for the UCSC Genome Browser and contains at least one full-length isoform for the vast majority of expressed genes in each tissue.
Collapse
Affiliation(s)
- Matthew Adams
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California 95064, USA
| | - Christopher Vollmers
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, California 95064, USA
| |
Collapse
|
28
|
Bilgrav Saether K, Eisfeldt J, Bengtsson JD, Lun MY, Grochowski CM, Mahmoud M, Chao HT, Rosenfeld JA, Liu P, Ek M, Schuy J, Ameur A, Dai H, Hwang JP, Sedlazeck FJ, Bi W, Marom R, Wincent J, Nordgren A, Carvalho CMB, Lindstrand A. Leveraging the T2T assembly to resolve rare and pathogenic inversions in reference genome gaps. Genome Res 2024; 34:1785-1797. [PMID: 39486878 DOI: 10.1101/gr.279346.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/12/2024] [Indexed: 11/04/2024]
Abstract
Chromosomal inversions (INVs) are particularly challenging to detect due to their copy-number neutral state and association with repetitive regions. Inversions represent about 1/20 of all balanced structural chromosome aberrations and can lead to disease by gene disruption or altering regulatory regions of dosage-sensitive genes in cis Short-read genome sequencing (srGS) can only resolve ∼70% of cytogenetically visible inversions referred to clinical diagnostic laboratories, likely due to breakpoints in repetitive regions. Here, we study 12 inversions by long-read genome sequencing (lrGS) (n = 9) or srGS (n = 3) and resolve nine of them. In four cases, the inversion breakpoint region was missing from at least one of the human reference genomes (GRCh37, GRCh38, T2T-CHM13) and a reference agnostic analysis was needed. One of these cases, an INV9 mappable only in de novo assembled lrGS data using T2T-CHM13 disrupts EHMT1 consistent with a Mendelian diagnosis (Kleefstra syndrome 1; MIM#610253). Next, by pairwise comparison between T2T-CHM13, GRCh37, and GRCh38, as well as the chimpanzee and bonobo, we show that hundreds of megabases of sequence are missing from at least one human reference, highlighting that primate genomes contribute to genomic diversity. Aligning population genomic data to these regions indicated that these regions are variable between individuals. Our analysis emphasizes that T2T-CHM13 is necessary to maximize the value of lrGS for optimal inversion detection in clinical diagnostics. These results highlight the importance of leveraging diverse and comprehensive reference genomes to resolve unsolved molecular cases in rare diseases.
Collapse
Affiliation(s)
- Kristine Bilgrav Saether
- Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76 Stockholm, Sweden
- Science for Life Laboratory, Karolinska Insitutet, 171 65 Solna, Sweden
| | - Jesper Eisfeldt
- Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76 Stockholm, Sweden;
- Science for Life Laboratory, Karolinska Insitutet, 171 65 Solna, Sweden
- Department of Clinical Genetics and Genomics, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Jesse D Bengtsson
- Pacific Northwest Research Institute, Seattle, Washington 98122, USA
| | - Ming Yin Lun
- Pacific Northwest Research Institute, Seattle, Washington 98122, USA
| | - Christopher M Grochowski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Medhat Mahmoud
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Center for Precision Health, McWilliams School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Hsiao-Tuan Chao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Texas Children's Hospital, Houston, Texas 77030, USA
- Cain Pediatric Neurology Research Laboratories, Jan and Dan Duncan Neurological Research Institute, Houston, Texas 77030, USA
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030, USA
- McNair Medical Institute, The Robert and Janice McNair Foundation, Houston, Texas 77024, USA
| | - Jill A Rosenfeld
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Pengfei Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Baylor Genetics Laboratory, Baylor College of Medicine, Houston, Texas 77021, USA
| | - Marlene Ek
- Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76 Stockholm, Sweden
- Department of Clinical Genetics and Genomics, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Jakob Schuy
- Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Adam Ameur
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Hongzheng Dai
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Baylor Genetics Laboratory, Baylor College of Medicine, Houston, Texas 77021, USA
| | - James Paul Hwang
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Fritz J Sedlazeck
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Computer Science, Rice University, Houston, Texas 77251, USA
| | - Weimin Bi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Baylor Genetics Laboratory, Baylor College of Medicine, Houston, Texas 77021, USA
| | - Ronit Marom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
- Texas Children's Hospital, Houston, Texas 77030, USA
| | - Josephine Wincent
- Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76 Stockholm, Sweden
- Department of Clinical Genetics and Genomics, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Ann Nordgren
- Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76 Stockholm, Sweden
- Department of Clinical Genetics and Genomics, Karolinska University Hospital, 171 76 Stockholm, Sweden
- Department of Laboratory Medicine, University of Gothenburg, 413 45 Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
| | | | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Karolinska Institute, 171 76 Stockholm, Sweden;
- Department of Clinical Genetics and Genomics, Karolinska University Hospital, 171 76 Stockholm, Sweden
| |
Collapse
|
29
|
Jepsen WM, Fazenbaker A, Ramsey K, Bonfitto A, Naymik M, Turner B, Sloan J, Tiwari N, Bernes SM, Neilson DE, Sanchez-Castillo M, Huentelman MJ, Narayanan V. Duchenne Muscular Dystrophy in Two Half-Brothers Due to Inherited 306 Kb Inverted Insertion of 10p15.1 into Intron 44 of the Dp427m Transcript of the DMD Gene. Int J Mol Sci 2024; 25:11922. [PMID: 39595988 PMCID: PMC11593467 DOI: 10.3390/ijms252211922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare genetic disorder caused by the absence of a fully functional dystrophin protein in myocytes. In skeletal muscle, the lack of dystrophin ultimately results in muscle wasting and the replacement of myocytes with fatty or fibrous tissues. In the heart, cardiomyocytes eventually fail and cause fatal cardiomyopathy. We present a case of a male patient and his younger brother with a maternally inherited inverted insertion of approximately 306 kb of chromosome 10 in the deep intronic region between exons 44 and 45 of the DMD gene, leading to Duchenne muscular dystrophy. Chromosomal microarray, comprehensive muscular dystrophy genetic testing, and whole exome sequencing were negative. Targeted transcriptome RNA sequencing at an external lab showed no aberrant splicing. Research whole genome sequencing identified the copy number gain and insertion. Subsequent reanalysis of the RNA sequencing data showed possible aberrant splicing involving DMD exons 44-45, and research RNA sequencing revealed a fusion between the DMD gene on the minus strand of chromosome X and the PFKFB3 gene on the plus strand of chromosome 10. We demonstrate that whole genome sequencing can be valuable for identifying intronic events in the DMD gene previously undetected or not reported by traditional clinical testing.
Collapse
Affiliation(s)
- Wayne M. Jepsen
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | | | - Keri Ramsey
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Anna Bonfitto
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Marcus Naymik
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Bryce Turner
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Jennifer Sloan
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | | | | | | | - Meredith Sanchez-Castillo
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Matt J. Huentelman
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| | - Vinodh Narayanan
- Center for Rare Childhood Disorders (C4RCD), Translational Genomics Research Institute (TGen), Phoenix, AZ 85004, USA; (W.M.J.)
| |
Collapse
|
30
|
Bond A, Fiaz S, Rollins K, Nario JEQ, Snyder ET, Atkins DJ, Rosen SJ, Granados A, Dey SS, Wilson MZ, Morrissey MA. Prior Fc receptor activation primes macrophages for increased sensitivity to IgG via long-term and short-term mechanisms. Dev Cell 2024; 59:2882-2896.e7. [PMID: 39137774 PMCID: PMC11537821 DOI: 10.1016/j.devcel.2024.07.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/17/2024] [Accepted: 07/16/2024] [Indexed: 08/15/2024]
Abstract
Macrophages measure the "eat-me" signal immunoglobulin G (IgG) to identify targets for phagocytosis. We tested whether prior encounters with IgG influence macrophage appetite. IgG is recognized by the Fc receptor. To temporally control Fc receptor activation, we engineered an Fc receptor that is activated by the light-induced oligomerization of Cry2, triggering phagocytosis. Using this tool, we demonstrate that subthreshold Fc receptor activation primes mouse bone-marrow-derived macrophages to be more sensitive to IgG in future encounters. Macrophages that have previously experienced subthreshold Fc receptor activation eat more IgG-bound human cancer cells. Increased phagocytosis occurs by two discrete mechanisms-a short- and long-term priming. Long-term priming requires new protein synthesis and Erk activity. Short-term priming does not require new protein synthesis and correlates with an increase in Fc receptor mobility. Our work demonstrates that IgG primes macrophages for increased phagocytosis, suggesting that therapeutic antibodies may become more effective after initial priming doses.
Collapse
Affiliation(s)
- Annalise Bond
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Sareen Fiaz
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Kirstin Rollins
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Jazz Elaiza Q Nario
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Erika T Snyder
- Biomolecular Science and Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Dixon J Atkins
- Biomolecular Science and Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Samuel J Rosen
- Biomolecular Science and Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Alyssa Granados
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Siddharth S Dey
- Chemical Engineering Department, University of California, Santa Barbara, Santa Barbara, CA, USA; Bioengineering Department, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Maxwell Z Wilson
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Meghan A Morrissey
- Molecular Cellular and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, CA, USA.
| |
Collapse
|
31
|
Wang NK, Wiltsie N, Winata HK, Fitz-Gibbon S, Gonzalez AE, Zeltser N, Agrawal R, Oh J, Arbet J, Patel Y, Yamaguchi TN, Boutros PC. StableLift: Optimized Germline and Somatic Variant Detection Across Genome Builds. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.31.621401. [PMID: 39554127 PMCID: PMC11565985 DOI: 10.1101/2024.10.31.621401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Reference genomes are foundational to modern genomics. Our growing understanding of genome structure leads to continual improvements in reference genomes and new genome "builds" with incompatible coordinate systems. We quantified the impact of genome build on germline and somatic variant calling by analyzing tumour-normal whole-genome pairs against the two most widely used human genome builds. The average individual had a build-discordance of 3.8% for germline SNPs, 8.6% for germline SVs, 25.9% for somatic SNVs and 49.6% for somatic SVs. Build-discordant variants are not simply false-positives: 47% were verified by targeted resequencing. Build-discordant variants were associated with specific genomic and technical features in variant- and algorithm-specific patterns. We leveraged these patterns to create StableLift, an algorithm that predicts cross-build stability with AUROCs of 0.934 ± 0.029. These results call for significant caution in cross-build analyses and for use of StableLift as a computationally efficient solution to mitigate inter-build artifacts.
Collapse
Affiliation(s)
- Nicholas K. Wang
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Nicholas Wiltsie
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Helena K. Winata
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Sorel Fitz-Gibbon
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Alfredo E. Gonzalez
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Nicole Zeltser
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Raag Agrawal
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Jieun Oh
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Jaron Arbet
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
- Department of Urology, University of California, Los Angeles
| | - Yash Patel
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Takafumi N. Yamaguchi
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
| | - Paul C. Boutros
- Department of Human Genetics, University of California, Los Angeles
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles
- Institute for Precision Health, University of California, Los Angeles
- Department of Urology, University of California, Los Angeles
| |
Collapse
|
32
|
Werner B, Powell E, Duggan J, Cortesi M, Lee YC, Arora V, Athavale R, Dean M, Warton K, Ford CE. Cell-free DNA from ascites identifies clinically relevant variants and tumour evolution in patients with advanced ovarian cancer. Mol Oncol 2024; 18:2668-2683. [PMID: 39115191 PMCID: PMC11547227 DOI: 10.1002/1878-0261.13710] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 07/08/2024] [Accepted: 07/23/2024] [Indexed: 11/09/2024] Open
Abstract
The emergence of targeted therapies has transformed ovarian cancer treatment. However, biomarker profiling for precision medicine is limited by access to quality, tumour-enriched tissue samples. The use of cell-free DNA (cfDNA) in ascites presents a potential solution to this challenge. In this study, next-generation sequencing was performed on ascites-derived cfDNA samples (26 samples from 15 human participants with ovarian cancer), with matched DNA from ascites-derived tumour cells (n = 5) and archived formalin-fixed paraffin-embedded (FFPE) tissue (n = 5). Similar tumour purity and variant detection were achieved with cfDNA compared to FFPE and ascites cell DNA. Analysis of large-scale genomic alterations, loss of heterozygosity and tumour mutation burden identified six cases of high genomic instability (including four with pathogenic BRCA1 and BRCA2 mutations). Copy number profiles and subclone prevalence changed between sequential ascites samples, particularly in a case where deletions and chromothripsis in Chr17p13.1 and Chr8q resulted in changes in clinically relevant TP53 and MYC variants over time. Ascites cfDNA identified clinically actionable information, concordant to tissue biopsies, enabling opportunistic molecular profiling. This advocates for analysis of ascites cfDNA in lieu of accessing tumour tissue via biopsy.
Collapse
Affiliation(s)
- Bonnita Werner
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyAustralia
| | - Elyse Powell
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyAustralia
| | - Jennifer Duggan
- Gynaecological Oncology DepartmentRoyal Hospital for WomenSydneyAustralia
| | - Marilisa Cortesi
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyAustralia
- Laboratory of Cellular and Molecular Engineering, Department of Electrical, Electronic and Information EngineeringAlma Mater Studiorum‐University of BolognaItaly
| | - Yeh Chen Lee
- Gynaecological Oncology DepartmentRoyal Hospital for WomenSydneyAustralia
- School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyAustralia
| | - Vivek Arora
- School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyAustralia
- Prince of Wales Private HospitalSydneyAustralia
| | - Ramanand Athavale
- Gynaecological Oncology DepartmentRoyal Hospital for WomenSydneyAustralia
- School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyAustralia
| | - Michael Dean
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and GeneticsNational Cancer InstituteRockvilleMDUSA
| | - Kristina Warton
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyAustralia
| | - Caroline E. Ford
- Gynaecological Cancer Research Group, School of Clinical Medicine, Faculty of Medicine and HealthUniversity of New South WalesSydneyAustralia
| |
Collapse
|
33
|
Cheung K, Rollins LA, Hammond JM, Barton K, Ferguson JM, Eyck HJF, Shine R, Edwards RJ. Repeat-Rich Regions Cause False-Positive Detection of NUMTs: A Case Study in Amphibians Using an Improved Cane Toad Reference Genome. Genome Biol Evol 2024; 16:evae246. [PMID: 39548850 PMCID: PMC11606642 DOI: 10.1093/gbe/evae246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/08/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
Mitochondrial DNA (mtDNA) has been widely used in genetics research for decades. Contamination from nuclear DNA of mitochondrial origin (NUMTs) can confound studies of phylogenetic relationships and mtDNA heteroplasmy. Homology searches with mtDNA are widely used to detect NUMTs in the nuclear genome. Nevertheless, false-positive detection of NUMTs is common when handling repeat-rich sequences, while fragmented genomes might result in missing true NUMTs. In this study, we investigated different NUMT detection methods and how the quality of the genome assembly affects them. We presented an improved nuclear genome assembly (aRhiMar1.3) of the invasive cane toad (Rhinella marina) with additional long-read Nanopore and 10× linked-read sequencing. The final assembly was 3.47 Gb in length with 91.3% of tetrapod universal single-copy orthologs (n = 5,310), indicating the gene-containing regions were well assembled. We used 3 complementary methods (NUMTFinder, dinumt, and PALMER) to study the NUMT landscape of the cane toad genome. All 3 methods yielded consistent results, showing very few NUMTs in the cane toad genome. Furthermore, we expanded NUMT detection analyses to other amphibians and confirmed a weak relationship between genome size and the number of NUMTs present in the nuclear genome. Amphibians are repeat-rich, and we show that the number of NUMTs found in highly repetitive genomes is prone to inflation when using homology-based detection without filters. Together, this study provides an exemplar of how to robustly identify NUMTs in complex genomes when confounding effects on mtDNA analyses are a concern.
Collapse
Affiliation(s)
- Kelton Cheung
- Evolution & Ecology Research Centre, School of Biological, Earth & Environmental Sciences, University of New South Wales, Sydney, Australia
- Evolution & Ecology Research Centre, School of Biotechnology & Biomolecular Sciences, University of New South Wales, Sydney, Australia
| | - Lee Ann Rollins
- Evolution & Ecology Research Centre, School of Biological, Earth & Environmental Sciences, University of New South Wales, Sydney, Australia
| | - Jillian M Hammond
- Genomics and Inherited Disease Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Centre for Population Genomics, Garvan Institute of Medical Research and Murdoch Children's Research Institute, Darlinghurst, New South Wales, Australia
| | - Kirston Barton
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - James M Ferguson
- Centre for Population Genomics, Garvan Institute of Medical Research and Murdoch Children's Research Institute, Darlinghurst, New South Wales, Australia
| | - Harrison J F Eyck
- National Collections and Marine Infrastructure, CSIRO, Canberra, Australian Capital Territory, Australia
| | - Richard Shine
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Richard J Edwards
- Evolution & Ecology Research Centre, School of Biotechnology & Biomolecular Sciences, University of New South Wales, Sydney, Australia
- Minderoo OceanOmics Centre at UWA, Oceans Institute, The University of Western Australia, Western Australia, Australia
| |
Collapse
|
34
|
Chang L, Xie Y, Taylor B, Wang Z, Sun J, Armand EJ, Mishra S, Xu J, Tastemel M, Lie A, Gibbs ZA, Indralingam HS, Tan TM, Bejar R, Chen CC, Furnari FB, Hu M, Ren B. Droplet Hi-C enables scalable, single-cell profiling of chromatin architecture in heterogeneous tissues. Nat Biotechnol 2024:10.1038/s41587-024-02447-1. [PMID: 39424717 DOI: 10.1038/s41587-024-02447-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
Current methods for analyzing chromatin architecture are not readily scalable to heterogeneous tissues. Here we introduce Droplet Hi-C, which uses a commercial microfluidic device for high-throughput, single-cell chromatin conformation profiling in droplets. Using Droplet Hi-C, we mapped the chromatin architecture of the mouse cortex and analyzed gene regulatory programs in major cortical cell types. In addition, we used this technique to detect copy number variations, structural variations and extrachromosomal DNA in human glioblastoma, colorectal and blood cancer cells, revealing clonal dynamics and other oncogenic events during treatment. We refined the technique to allow joint profiling of chromatin architecture and transcriptome in single cells, facilitating exploration of the links between chromatin architecture and gene expression in both normal tissues and tumors. Thus, Droplet Hi-C both addresses critical gaps in chromatin analysis of heterogeneous tissues and enhances understanding of gene regulation.
Collapse
Affiliation(s)
- Lei Chang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Yang Xie
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Brett Taylor
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
- Medical Scientist Training Program, University of California, San Diego, La Jolla, CA, USA
| | - Zhaoning Wang
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jiachen Sun
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Systems Biology and Bioinformatics PhD Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ethan J Armand
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California, San Diego, La Jolla, CA, USA
| | - Shreya Mishra
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Jie Xu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Melodi Tastemel
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Audrey Lie
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Zane A Gibbs
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Hannah S Indralingam
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Tuyet M Tan
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Rafael Bejar
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, USA
| | - Frank B Furnari
- Department of Medicine, University of California, San Diego School of Medicine, La Jolla, CA, USA
| | - Ming Hu
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Bing Ren
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA.
- Moores Cancer Center, UC San Diego, La Jolla, CA, USA.
- Center for Epigenomics, Institute for Genomic Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
35
|
Mishra R, Thunuguntla P, Perkin A, Duraiyan D, Bagwill K, Gonzales S, Brizuela V, Daly S, Chang YJ, Abebe M, Rajana Y, Wichmann K, Bolick C, King J, Fiala M, Fortier J, Jayasinghe R, Schroeder M, Ding L, Vij R, Silva-Fisher J. LINC01432 binds to CELF2 in newly diagnosed multiple myeloma promoting short progression-free survival to standard therapy. RESEARCH SQUARE 2024:rs.3.rs-4888379. [PMID: 39483883 PMCID: PMC11527149 DOI: 10.21203/rs.3.rs-4888379/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Multiple Myeloma (MM) is an incurable form of cancer that arises from malignant plasma cells, with over 35,000 new cases diagnosed annually in the United States. While there are a growing number of approved therapies, MM remains incurable and nearly all patients will relapse and exhaust available treatments. Mechanisms for disease progression are unclear and little is known regarding the role of long non-coding RNAs (lncRNA) in mediating disease progression and response to treatment. Here, we used transcriptome sequencing to compare newly diagnosed MM (NDMM) patients who had short progression-free survival (PFS) to standard first-line treatment (PFS < 24 months) to patients who had prolonged PFS (PFS > 24 months). We identified 157 differentially upregulated lncRNAs with short PFS and focused our efforts on characterizing the most upregulated lncRNA, LINC01432. We investigated LINC01432 to show that its overexpression significantly increases cell viability and reduces apoptosis, while knockdown significantly reduces viability and increases apoptosis. Next, we show that LINC01432 directly interacts with the RNA binding protein, CELF2. Lastly, we showed that LINC01432-targeted locked nucleic acid antisense oligonucleotides reduce viability and increases apoptosis. In summary, this fundamental study identified lncRNAs associated with short PFS to standard NDMM treatment and further characterized LINC01432.
Collapse
Affiliation(s)
- Richa Mishra
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Prasanth Thunuguntla
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Alani Perkin
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Dhanusha Duraiyan
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Katelyn Bagwill
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Savannah Gonzales
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Vanessa Brizuela
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Steve Daly
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Yoon Jae Chang
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Mahdote Abebe
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Yash Rajana
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Kelly Wichmann
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Catheryn Bolick
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Jaiyana King
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Mark Fiala
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| | - Julie Fortier
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Reyka Jayasinghe
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
| | - Mark Schroeder
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| | - Li Ding
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| | - Ravi Vij
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| | - Jessica Silva-Fisher
- Department of Internal Medicine, Division of Oncology, School of Medicine, Washington University in St. Louis, MO, 631101
- Siteman Cancer Center, Washington University in St. Louis, MO, 631102
| |
Collapse
|
36
|
Henglin M, Ghareghani M, Harvey WT, Porubsky D, Koren S, Eichler EE, Ebert P, Marschall T. Graphasing: phasing diploid genome assembly graphs with single-cell strand sequencing. Genome Biol 2024; 25:265. [PMID: 39390579 PMCID: PMC11466045 DOI: 10.1186/s13059-024-03409-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 09/30/2024] [Indexed: 10/12/2024] Open
Abstract
Haplotype information is crucial for biomedical and population genetics research. However, current strategies to produce de novo haplotype-resolved assemblies often require either difficult-to-acquire parental data or an intermediate haplotype-collapsed assembly. Here, we present Graphasing, a workflow which synthesizes the global phase signal of Strand-seq with assembly graph topology to produce chromosome-scale de novo haplotypes for diploid genomes. Graphasing readily integrates with any assembly workflow that both outputs an assembly graph and has a haplotype assembly mode. Graphasing performs comparably to trio phasing in contiguity, phasing accuracy, and assembly quality, outperforms Hi-C in phasing accuracy, and generates human assemblies with over 18 chromosome-spanning haplotypes.
Collapse
Affiliation(s)
- Mir Henglin
- Institute for Medical Biometry and Bioinformatics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Center for Digital Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maryam Ghareghani
- Department of Mathematics and Computer Science, Freie Universität Berlin, Berlin, Germany
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - William T Harvey
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - David Porubsky
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
| | - Sergey Koren
- Genome Informatics Section, Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Evan E Eichler
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Peter Ebert
- Institute for Medical Biometry and Bioinformatics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Center for Digital Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Core Unit Bioinformatics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| | - Tobias Marschall
- Institute for Medical Biometry and Bioinformatics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Center for Digital Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
37
|
Gaye A, Sene ARG, Gadji M, Deme A, Cisse A, Ndiaye R. Toward building a comprehensive human pan-genome: The SEN-GENOME project. Am J Hum Genet 2024; 111:2074-2078. [PMID: 39305906 PMCID: PMC11480787 DOI: 10.1016/j.ajhg.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/22/2024] [Accepted: 08/22/2024] [Indexed: 10/06/2024] Open
Abstract
The human reference genome (GRCh38), primarily sourced from individuals of European descent, falls short in capturing the vast genetic diversity across global populations. Efforts to diversify the reference genome face challenges in accessibility and representation, exacerbating the scarcity of African genomic data crucial for studying diseases prevalent in these populations. Sherman et al. proposed constructing reference genomes tailored to distinct human sub-populations. Their African Pan-Genome initiative highlighted substantial genetic variation missing from the GRCh38 human reference genome, emphasizing the necessity for population-specific genomes. In response, local initiatives like the Senegalese Genome project (SEN-GENOME) have emerged to document the genomes of historically overlooked populations. SEN-GENOME embodies community-driven decentralized research. With meticulous recruitment criteria and ethical practices, it aims to sequence 1,000 genomes from 31 ethnolinguistic groups, in the fourteen administrative regions of Senegal, fostering local genomic research tailored to the region. The key to SEN-GENOME's success is its commitment to local governance of data, capacity building, and integration with broader pan-genome projects in Africa. Despite the complexities of data harmonization and sharing, our collaborative efforts are aligned with common goals, ensuring steady progress toward a comprehensive human pan-genome. We invite and welcome collaboration with other research entities to achieve this shared vision. In summary, local initiatives such as SEN-GENOME are pivotal in bridging genomic disparities, offering pathways to equitable and inclusive genomic research. Collaborative endeavors guided by a collective vision for human health will propel us toward a more encompassing understanding of the human genome and better health through genomic medicine.
Collapse
Affiliation(s)
- Amadou Gaye
- Department of Integrative Genomics and Epidemiology, School of Graduate Studies, Meharry Medical College, Nashville, TN, USA.
| | - Andrea Regina G Sene
- Division of Human Genetics, Faculty of Medicine, Pharmacy and Odontology, University Cheikh Anta Diop, Dakar, Senegal
| | - Macoura Gadji
- Service of Biological Hematology & Oncology-Hematology, Faculty of Medicine, Pharmacy and Odontology Stomatology, University Cheikh Anta Diop, Dakar, Senegal
| | - Alioune Deme
- Laboratory of Prehistory and Cultural Heritage, Department of History, Faculty of Arts and Humanities, University Cheikh Anta Diop, Dakar, Senegal
| | - Aynina Cisse
- Senegalese National Academy of Science and Technology, Dakar, Senegal
| | - Rokhaya Ndiaye
- Division of Human Genetics, Faculty of Medicine, Pharmacy and Odontology, University Cheikh Anta Diop, Dakar, Senegal.
| |
Collapse
|
38
|
Hung TK, Liu WC, Lai SK, Chuang HW, Lee YC, Lin HY, Hsu CL, Chen CY, Yang YC, Hsu JS, Chen PL. Genetic complexity of killer-cell immunoglobulin-like receptor genes in human pangenome assemblies. Genome Res 2024; 34:1211-1223. [PMID: 39251346 PMCID: PMC11444179 DOI: 10.1101/gr.278358.123] [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: 11/12/2023] [Accepted: 08/14/2024] [Indexed: 09/11/2024]
Abstract
The killer-cell immunoglobulin-like receptor (KIR) gene complex, a highly polymorphic region of the human genome that encodes proteins involved in immune responses, poses strong challenges in genotyping owing to its remarkable genetic diversity and structural intricacy. Accurate analysis of KIR alleles, including their structural variations, is crucial for understanding their roles in various immune responses. Leveraging the high-quality genome assemblies from the Human Pangenome Reference Consortium (HPRC), we present a novel bioinformatic tool, the structural KIR annoTator (SKIRT), to investigate gene diversity and facilitate precise KIR allele analysis. In 47 HPRC-phased assemblies, SKIRT identifies a recurrent novel KIR2DS4/3DL1 fusion gene in the paternal haplotype of HG02630 and maternal haplotype of NA19240. Additionally, SKIRT accurately identifies eight structural variants and 15 novel nonsynonymous alleles, all of which are independently validated using short-read data or quantitative polymerase chain reaction. Our study has discovered a total of 570 novel alleles, among which eight haplotypes harbor at least one KIR gene duplication, six haplotypes have lost at least one framework gene, and 75 out of 94 haplotypes (79.8%) carry at least five novel alleles, thus confirming KIR genetic diversity. These findings are pivotal in providing insights into KIR gene diversity and serve as a solid foundation for understanding the functional consequences of KIR structural variations. High-resolution genome assemblies offer unprecedented opportunities to explore polymorphic regions that are challenging to investigate using short-read sequencing methods. The SKIRT pipeline emerges as a highly efficient tool, enabling the comprehensive detection of the complete spectrum of KIR alleles within human genome assemblies.
Collapse
Affiliation(s)
- Tsung-Kai Hung
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
| | - Wan-Chi Liu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100229, Taiwan
| | - Sheng-Kai Lai
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100229, Taiwan
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei 10617, Taiwan
| | - Hui-Wen Chuang
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
| | - Yi-Che Lee
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100229, Taiwan
| | - Hong-Ye Lin
- Department of Biomechatronics Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Chia-Lang Hsu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Taipei 100229, Taiwan
| | - Chien-Yu Chen
- Department of Biomechatronics Engineering, National Taiwan University, Taipei 10617, Taiwan
| | - Ya-Chien Yang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei 100229, Taiwan;
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei 100229, Taiwan
| | - Jacob Shujui Hsu
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100233, Taiwan;
| | - Pei-Lung Chen
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei 100233, Taiwan;
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 100229, Taiwan
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei 10617, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100229, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, National Taiwan University Hospital, Taipei 100229, Taiwan
| |
Collapse
|
39
|
Moon N, Morgan CP, Marx-Rattner R, Jeng A, Johnson RL, Chikezie I, Mannella C, Sammel MD, Epperson CN, Bale TL. Stress increases sperm respiration and motility in mice and men. Nat Commun 2024; 15:7900. [PMID: 39261485 PMCID: PMC11391062 DOI: 10.1038/s41467-024-52319-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
Semen quality and fertility has declined over the last 50 years, corresponding to ever-increasing environmental stressors. However, the cellular mechanisms involved and their impact on sperm functions remain unknown. In a repeated sampling human cohort study, we identify a significant effect of prior perceived stress to increase sperm motility 2-3 months following stress, timing that expands upon our previous studies revealing significant stress-associated changes in sperm RNA important for fertility. We mechanistically examine this post-stress timing in mice using an in vitro stress model in the epididymal epithelial cells responsible for sperm maturation and find 7282 differentially H3K27me3 bound DNA regions involving genes critical for mitochondrial and metabolic pathways. Further, prior stress exposure significantly changes the composition and size of epithelial cell-secreted extracellular vesicles that when incubated with mouse sperm, increase mitochondrial respiration and sperm motility, adding to our prior work showing impacts on embryo development. Together, these studies identify a time-dependent, translational signaling pathway that communicates stress experience to sperm, ultimately affecting reproductive functions.
Collapse
Affiliation(s)
- Nickole Moon
- Department of Psychiatry, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Christopher P Morgan
- Department of Pharmacology, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Ruth Marx-Rattner
- Department of Pharmacology, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Alyssa Jeng
- Department of Psychiatry, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, 80045, USA
| | - Rachel L Johnson
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ijeoma Chikezie
- Department of Pharmacology, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Carmen Mannella
- Department of Physiology, University of Maryland Baltimore, Baltimore, MD, 21201, USA
| | - Mary D Sammel
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - C Neill Epperson
- Department of Psychiatry, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, 80045, USA
| | - Tracy L Bale
- Department of Psychiatry, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Maryland Baltimore, Baltimore, MD, 21201, USA.
| |
Collapse
|
40
|
Carlisle SG, Albasha H, Michelena HI, Sabate-Rotes A, Bianco L, De Backer J, Mosquera LM, Yetman AT, Bissell MM, Andreassi MG, Foffa I, Hui DS, Caffarelli A, Kim YY, Guo D, Citro R, De Marco M, Tretter JT, McBride KL, Milewicz DM, Body SC, Prakash SK. Rare genomic copy number variants implicate new candidate genes for bicuspid aortic valve. PLoS One 2024; 19:e0304514. [PMID: 39240962 PMCID: PMC11379187 DOI: 10.1371/journal.pone.0304514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/14/2024] [Indexed: 09/08/2024] Open
Abstract
Bicuspid aortic valve (BAV), the most common congenital heart defect, is a major cause of aortic valve disease requiring valve interventions and thoracic aortic aneurysms predisposing to acute aortic dissections. The spectrum of BAV ranges from early onset valve and aortic complications (EBAV) to sporadic late onset disease. Rare genomic copy number variants (CNVs) have previously been implicated in the development of BAV and thoracic aortic aneurysms. We determined the frequency and gene content of rare CNVs in EBAV probands (n = 272) using genome-wide SNP microarray analysis and three complementary CNV detection algorithms (cnvPartition, PennCNV, and QuantiSNP). Unselected control genotypes from the Database of Genotypes and Phenotypes were analyzed using identical methods. We filtered the data to select large genic CNVs that were detected by multiple algorithms. Findings were replicated in a BAV cohort with late onset sporadic disease (n = 5040). We identified 3 large and rare (< 1,1000 in controls) CNVs in EBAV probands. The burden of CNVs intersecting with genes known to cause BAV when mutated was increased in case-control analysis. CNVs intersecting with GATA4 and DSCAM were enriched in cases, recurrent in other datasets, and segregated with disease in families. In total, we identified potentially pathogenic CNVs in 9% of EBAV cases, implicating alterations of candidate genes at these loci in the pathogenesis of BAV.
Collapse
Affiliation(s)
- Steven G Carlisle
- University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Hasan Albasha
- University College Dublin School of Medicine, Dublin, Ireland
| | | | | | - Lisa Bianco
- Vall d'Hebron University Hospital, Barcelona, Spain
| | | | | | - Anji T Yetman
- University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | | | | | - Ilenia Foffa
- Consiglio Nazionale delle Richerche (CNR), Instituto di Fisiologia Clinica, Pisa, Italy
| | - Dawn S Hui
- University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States of America
| | - Anthony Caffarelli
- Hoag Memorial Hospital Presbyterian, Newport Beach, California, United States of America
| | - Yuli Y Kim
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Dongchuan Guo
- University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Rodolfo Citro
- University Hospital "San Giovanni di Dio e Ruggi d'Aragona," Salerno, Italy
| | - Margot De Marco
- Schola Medica Salernitana, University of Salerno, Baronissi, Italy
| | | | - Kim L McBride
- University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Dianna M Milewicz
- University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Simon C Body
- Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Siddharth K Prakash
- University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| |
Collapse
|
41
|
DeGroat W, Inoue F, Ashuach T, Yosef N, Ahituv N, Kreimer A. Comprehensive network modeling approaches unravel dynamic enhancer-promoter interactions across neural differentiation. Genome Biol 2024; 25:221. [PMID: 39143563 PMCID: PMC11323586 DOI: 10.1186/s13059-024-03365-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/01/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Increasing evidence suggests that a substantial proportion of disease-associated mutations occur in enhancers, regions of non-coding DNA essential to gene regulation. Understanding the structures and mechanisms of the regulatory programs this variation affects can shed light on the apparatuses of human diseases. RESULTS We collect epigenetic and gene expression datasets from seven early time points during neural differentiation. Focusing on this model system, we construct networks of enhancer-promoter interactions, each at an individual stage of neural induction. These networks serve as the base for a rich series of analyses, through which we demonstrate their temporal dynamics and enrichment for various disease-associated variants. We apply the Girvan-Newman clustering algorithm to these networks to reveal biologically relevant substructures of regulation. Additionally, we demonstrate methods to validate predicted enhancer-promoter interactions using transcription factor overexpression and massively parallel reporter assays. CONCLUSIONS Our findings suggest a generalizable framework for exploring gene regulatory programs and their dynamics across developmental processes; this includes a comprehensive approach to studying the effects of disease-associated variation on transcriptional networks. The techniques applied to our networks have been published alongside our findings as a computational tool, E-P-INAnalyzer. Our procedure can be utilized across different cellular contexts and disorders.
Collapse
Affiliation(s)
- William DeGroat
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, 679 Hoes Lane West, Piscataway, NJ, 08854, USA
| | - Fumitaka Inoue
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Tal Ashuach
- Department of Electrical Engineering and Computer Sciences and Center for Computational Biology, University of California, Berkeley, 387 Soda Hall, Berkeley, CA, 94720, USA
| | - Nir Yosef
- Department of Systems Immunology, Weizmann Institute of Science, 234 Herzl Street, Rehovot, 7610001, Israel
- Chan-Zuckerberg Biohub, 499 Illinois St, San Francisco, CA, 94158, USA
- Department of Systems Immunology, Ragon Institute of MGH, MIT, and Harvard Institute of Science, 400 Technology Square, Cambridge, MA, 02139, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, 513 Parnassus Ave, San Francisco, CA, 94143, USA
- Institute for Human Genetics, University of California, 513 Parnassus Ave, San Francisco, CA, 94143, USA
| | - Anat Kreimer
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, 679 Hoes Lane West, Piscataway, NJ, 08854, USA.
- Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ, 08854, USA.
| |
Collapse
|
42
|
Taylor DJ, Eizenga JM, Li Q, Das A, Jenike KM, Kenny EE, Miga KH, Monlong J, McCoy RC, Paten B, Schatz MC. Beyond the Human Genome Project: The Age of Complete Human Genome Sequences and Pangenome References. Annu Rev Genomics Hum Genet 2024; 25:77-104. [PMID: 38663087 PMCID: PMC11451085 DOI: 10.1146/annurev-genom-021623-081639] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
The Human Genome Project was an enormous accomplishment, providing a foundation for countless explorations into the genetics and genomics of the human species. Yet for many years, the human genome reference sequence remained incomplete and lacked representation of human genetic diversity. Recently, two major advances have emerged to address these shortcomings: complete gap-free human genome sequences, such as the one developed by the Telomere-to-Telomere Consortium, and high-quality pangenomes, such as the one developed by the Human Pangenome Reference Consortium. Facilitated by advances in long-read DNA sequencing and genome assembly algorithms, complete human genome sequences resolve regions that have been historically difficult to sequence, including centromeres, telomeres, and segmental duplications. In parallel, pangenomes capture the extensive genetic diversity across populations worldwide. Together, these advances usher in a new era of genomics research, enhancing the accuracy of genomic analysis, paving the path for precision medicine, and contributing to deeper insights into human biology.
Collapse
Affiliation(s)
- Dylan J Taylor
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA; , ,
| | - Jordan M Eizenga
- Genomics Institute, University of California, Santa Cruz, California, USA; , ,
| | - Qiuhui Li
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland, USA; ,
| | - Arun Das
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland, USA; ,
| | - Katharine M Jenike
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA;
| | - Eimear E Kenny
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA;
| | - Karen H Miga
- Department of Biomolecular Engineering, University of California, Santa Cruz, California, USA
- Genomics Institute, University of California, Santa Cruz, California, USA; , ,
| | - Jean Monlong
- Institut de Recherche en Santé Digestive, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France;
| | - Rajiv C McCoy
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA; , ,
| | - Benedict Paten
- Department of Biomolecular Engineering, University of California, Santa Cruz, California, USA
- Genomics Institute, University of California, Santa Cruz, California, USA; , ,
| | - Michael C Schatz
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland, USA; ,
- Department of Biology, Johns Hopkins University, Baltimore, Maryland, USA; , ,
| |
Collapse
|
43
|
Talbot BM, Clennon JA, Rakotoarison MFN, Rautman L, Durry S, Ragazzo LJ, Wright PC, Gillespie TR, Read TD. Metagenome-wide characterization of shared antimicrobial resistance genes in sympatric people and lemurs in rural Madagascar. PeerJ 2024; 12:e17805. [PMID: 39099658 PMCID: PMC11296303 DOI: 10.7717/peerj.17805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/03/2024] [Indexed: 08/06/2024] Open
Abstract
Background Tracking the spread of antibiotic resistant bacteria is critical to reduce global morbidity and mortality associated with human and animal infections. There is a need to understand the role that wild animals in maintenance and transfer of antibiotic resistance genes (ARGs). Methods This study used metagenomics to identify and compare the abundance of bacterial species and ARGs detected in the gut microbiomes from sympatric humans and wild mouse lemurs in a forest-dominated, roadless region of Madagascar near Ranomafana National Park. We examined the contribution of human geographic location toward differences in ARG abundance and compared the genomic similarity of ARGs between host source microbiomes. Results Alpha and beta diversity of species and ARGs between host sources were distinct but maintained a similar number of detectable ARG alleles. Humans were differentially more abundant for four distinct tetracycline resistance-associated genes compared to lemurs. There was no significant difference in human ARG diversity from different locations. Human and lemur microbiomes shared 14 distinct ARGs with highly conserved in nucleotide identity. Synteny of ARG-associated assemblies revealed a distinct multidrug-resistant gene cassette carrying dfrA1 and aadA1 present in human and lemur microbiomes without evidence of geographic overlap, suggesting that these resistance genes could be widespread in this ecosystem. Further investigation into intermediary processes that maintain drug-resistant bacteria in wildlife settings is needed.
Collapse
Affiliation(s)
- Brooke M. Talbot
- Program in Population Biology, Ecology, and Evolution, Emory University, Atlanta, GA, United States of America
- Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA, United States of America
| | - Julie A. Clennon
- Department of Environmental Sciences, Emory University, Atlanta, GA, United States of America
| | | | - Lydia Rautman
- Department of Environmental Sciences, Emory University, Atlanta, GA, United States of America
| | - Sarah Durry
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - Leo J. Ragazzo
- Department of Environmental Sciences, Emory University, Atlanta, GA, United States of America
| | - Patricia C. Wright
- Centre ValBio, Ranomafana, Madagascar
- Institute for the Conservation of Tropical Ecosystems, State University of New York at Stony Brook, Stony Brook, NY, United States of America
| | - Thomas R. Gillespie
- Program in Population Biology, Ecology, and Evolution, Emory University, Atlanta, GA, United States of America
- Department of Environmental Sciences, Emory University, Atlanta, GA, United States of America
- Centre ValBio, Ranomafana, Madagascar
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States of America
| | - Timothy D. Read
- Program in Population Biology, Ecology, and Evolution, Emory University, Atlanta, GA, United States of America
- Division of Infectious Diseases, School of Medicine, Emory University, Atlanta, GA, United States of America
| |
Collapse
|
44
|
Lin MJ, Langmead B, Safonova Y. IGLoo: Profiling the Immunoglobulin Heavy chain locus in Lymphoblastoid Cell Lines with PacBio High-Fidelity Sequencing reads. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.20.604421. [PMID: 39091872 PMCID: PMC11291057 DOI: 10.1101/2024.07.20.604421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
New high-quality human genome assemblies derived from lymphoblastoid cell lines (LCLs) provide reference genomes and pangenomes for genomics studies. However, the characteristics of LCLs pose technical challenges to profiling immunoglobulin (IG) genes. IG loci in LCLs contain a mixture of germline and somatically recombined haplotypes, making them difficult to genotype or assemble accurately. To address these challenges, we introduce IGLoo, a software tool that implements novel methods for analyzing sequence data and genome assemblies derived from LCLs. IGLoo characterizes somatic V(D)J recombination events in the sequence data and identifies the breakpoints and missing IG genes in the LCL-based assemblies. Furthermore, IGLoo implements a novel reassembly framework to improve germline assembly quality by integrating information about somatic events and population structural variantions in the IG loci. We applied IGLoo to study the assemblies from the Human Pangenome Reference Consortium, providing new insights into the mechanisms, gene usage, and patterns of V(D)J recombination, causes of assembly fragmentation in the IG heavy chain (IGH) locus, and improved representation of the IGH assemblies.
Collapse
Affiliation(s)
- Mao-Jan Lin
- Department of Computer Science, Johns Hopkins University
| | - Ben Langmead
- Department of Computer Science, Johns Hopkins University
| | - Yana Safonova
- Department of Computer Science, Johns Hopkins University
- Computer Science and Engineering Department, Pennsylvania State University
- Huck Institutes of Life Sciences, Pennsylvania State University
| |
Collapse
|
45
|
Ungar RA, Goddard PC, Jensen TD, Degalez F, Smith KS, Jin CA, Bonner DE, Bernstein JA, Wheeler MT, Montgomery SB. Impact of genome build on RNA-seq interpretation and diagnostics. Am J Hum Genet 2024; 111:1282-1300. [PMID: 38834072 PMCID: PMC11267525 DOI: 10.1016/j.ajhg.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Transcriptomics is a powerful tool for unraveling the molecular effects of genetic variants and disease diagnosis. Prior studies have demonstrated that choice of genome build impacts variant interpretation and diagnostic yield for genomic analyses. To identify the extent genome build also impacts transcriptomics analyses, we studied the effect of the hg19, hg38, and CHM13 genome builds on expression quantification and outlier detection in 386 rare disease and familial control samples from both the Undiagnosed Diseases Network and Genomics Research to Elucidate the Genetics of Rare Disease Consortium. Across six routinely collected biospecimens, 61% of quantified genes were not influenced by genome build. However, we identified 1,492 genes with build-dependent quantification, 3,377 genes with build-exclusive expression, and 9,077 genes with annotation-specific expression across six routinely collected biospecimens, including 566 clinically relevant and 512 known OMIM genes. Further, we demonstrate that between builds for a given gene, a larger difference in quantification is well correlated with a larger change in expression outlier calling. Combined, we provide a database of genes impacted by build choice and recommend that transcriptomics-guided analyses and diagnoses are cross referenced with these data for robustness.
Collapse
Affiliation(s)
- Rachel A Ungar
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Pagé C Goddard
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Tanner D Jensen
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | | | - Kevin S Smith
- Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA
| | - Christopher A Jin
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA
| | - Devon E Bonner
- Department of Pediatrics, School of Medicine, Stanford University, Stanford, CA, USA; Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, USA
| | - Jonathan A Bernstein
- Stanford Center for Undiagnosed Diseases, Stanford University, Stanford, CA, USA
| | - Matthew T Wheeler
- Department of Cardiovascular Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Stephen B Montgomery
- Department of Genetics, School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, School of Medicine, Stanford University, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
| |
Collapse
|
46
|
Sandoval-Velasco M, Dudchenko O, Rodríguez JA, Pérez Estrada C, Dehasque M, Fontsere C, Mak SST, Khan R, Contessoto VG, Oliveira Junior AB, Kalluchi A, Zubillaga Herrera BJ, Jeong J, Roy RP, Christopher I, Weisz D, Omer AD, Batra SS, Shamim MS, Durand NC, O'Connell B, Roca AL, Plikus MV, Kusliy MA, Romanenko SA, Lemskaya NA, Serdyukova NA, Modina SA, Perelman PL, Kizilova EA, Baiborodin SI, Rubtsov NB, Machol G, Rath K, Mahajan R, Kaur P, Gnirke A, Garcia-Treviño I, Coke R, Flanagan JP, Pletch K, Ruiz-Herrera A, Plotnikov V, Pavlov IS, Pavlova NI, Protopopov AV, Di Pierro M, Graphodatsky AS, Lander ES, Rowley MJ, Wolynes PG, Onuchic JN, Dalén L, Marti-Renom MA, Gilbert MTP, Aiden EL. Three-dimensional genome architecture persists in a 52,000-year-old woolly mammoth skin sample. Cell 2024; 187:3541-3562.e51. [PMID: 38996487 DOI: 10.1016/j.cell.2024.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 03/07/2024] [Accepted: 06/03/2024] [Indexed: 07/14/2024]
Abstract
Analyses of ancient DNA typically involve sequencing the surviving short oligonucleotides and aligning to genome assemblies from related, modern species. Here, we report that skin from a female woolly mammoth (†Mammuthus primigenius) that died 52,000 years ago retained its ancient genome architecture. We use PaleoHi-C to map chromatin contacts and assemble its genome, yielding 28 chromosome-length scaffolds. Chromosome territories, compartments, loops, Barr bodies, and inactive X chromosome (Xi) superdomains persist. The active and inactive genome compartments in mammoth skin more closely resemble Asian elephant skin than other elephant tissues. Our analyses uncover new biology. Differences in compartmentalization reveal genes whose transcription was potentially altered in mammoths vs. elephants. Mammoth Xi has a tetradic architecture, not bipartite like human and mouse. We hypothesize that, shortly after this mammoth's death, the sample spontaneously freeze-dried in the Siberian cold, leading to a glass transition that preserved subfossils of ancient chromosomes at nanometer scale.
Collapse
Affiliation(s)
| | - Olga Dudchenko
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA.
| | - Juan Antonio Rodríguez
- Center for Evolutionary Hologenomics, University of Copenhagen, DK-1353 Copenhagen, Denmark; Centre Nacional d'Anàlisi Genòmica, CNAG, 08028 Barcelona, Spain
| | - Cynthia Pérez Estrada
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA
| | - Marianne Dehasque
- Centre for Palaeogenetics, SE-106 91 Stockholm, Sweden; Department of Bioinformatics and Genetics, Swedish Museum of Natural History, 10405 Stockholm, Sweden; Department of Zoology, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Claudia Fontsere
- Center for Evolutionary Hologenomics, University of Copenhagen, DK-1353 Copenhagen, Denmark
| | - Sarah S T Mak
- Center for Evolutionary Hologenomics, University of Copenhagen, DK-1353 Copenhagen, Denmark
| | - Ruqayya Khan
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | - Achyuth Kalluchi
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bernardo J Zubillaga Herrera
- Department of Physics, Northeastern University, Boston, MA 02115, USA; Center for Theoretical Biological Physics, Northeastern University, Boston, MA 02215, USA
| | - Jiyun Jeong
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Renata P Roy
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA; Departments of Biology and Physics, Texas Southern University, Houston, TX 77004, USA
| | - Ishawnia Christopher
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - David Weisz
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Arina D Omer
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sanjit S Batra
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Muhammad S Shamim
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Neva C Durand
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brendan O'Connell
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA; Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR 97239, USA
| | - Alfred L Roca
- Department of Animal Sciences and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Mariya A Kusliy
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk 630090, Russia
| | | | - Natalya A Lemskaya
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk 630090, Russia
| | | | - Svetlana A Modina
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk 630090, Russia
| | - Polina L Perelman
- Institute of Molecular and Cellular Biology SB RAS, Novosibirsk 630090, Russia
| | - Elena A Kizilova
- Institute of Cytology and Genetics SB RAS, Novosibirsk 630090, Russia
| | | | - Nikolai B Rubtsov
- Institute of Cytology and Genetics SB RAS, Novosibirsk 630090, Russia
| | - Gur Machol
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Krisha Rath
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ragini Mahajan
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA; Department of Biosciences, Rice University, Houston, TX 77005, USA
| | - Parwinder Kaur
- UWA School of Agriculture and Environment, University of Western Australia, Perth, WA 6009, Australia
| | - Andreas Gnirke
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | | | - Rob Coke
- San Antonio Zoo, San Antonio, TX 78212, USA
| | | | | | - Aurora Ruiz-Herrera
- Departament de Biologia Cel·lular, Fisiologia i Immunologia and Genome Integrity and Instability Group, Institut de Biotecnologia i Biomedicina, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | | | | | - Naryya I Pavlova
- Institute of Biological Problems of Cryolitezone SB RAS, Yakutsk 677000, Russia
| | - Albert V Protopopov
- Academy of Sciences of Sakha Republic, Yakutsk 677000, Russia; North-Eastern Federal University, Yakutsk 677027, Russia
| | - Michele Di Pierro
- Department of Physics, Northeastern University, Boston, MA 02115, USA; Center for Theoretical Biological Physics, Northeastern University, Boston, MA 02215, USA
| | | | - Eric S Lander
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - M Jordan Rowley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Peter G Wolynes
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA; Department of Biosciences, Rice University, Houston, TX 77005, USA; Departments of Physics, Astronomy, & Chemistry, Rice University, Houston, TX 77005, USA
| | - José N Onuchic
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA; Department of Biosciences, Rice University, Houston, TX 77005, USA; Departments of Physics, Astronomy, & Chemistry, Rice University, Houston, TX 77005, USA
| | - Love Dalén
- Centre for Palaeogenetics, SE-106 91 Stockholm, Sweden; Department of Bioinformatics and Genetics, Swedish Museum of Natural History, 10405 Stockholm, Sweden; Department of Zoology, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Marc A Marti-Renom
- Centre Nacional d'Anàlisi Genòmica, CNAG, 08028 Barcelona, Spain; Centre for Genomic Regulation, The Barcelona Institute for Science and Technology, 08003 Barcelona, Spain; ICREA, 08010 Barcelona, Spain; Universitat Pompeu Fabra, 08002 Barcelona, Spain.
| | - M Thomas P Gilbert
- Center for Evolutionary Hologenomics, University of Copenhagen, DK-1353 Copenhagen, Denmark; University Museum NTNU, 7012 Trondheim, Norway.
| | - Erez Lieberman Aiden
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
47
|
Jimenez N, Norton T, Diadala G, Bell E, Valenti M, Farland LV, Mahnert N, Herbst-Kralovetz MM. Vaginal and rectal microbiome contribute to genital inflammation in chronic pelvic pain. BMC Med 2024; 22:283. [PMID: 38972981 PMCID: PMC11229265 DOI: 10.1186/s12916-024-03500-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Chronic pelvic pain (CPP) is a multifactorial syndrome that can substantially affect a patient's quality of life. Endometriosis is one cause of CPP, and alterations of the immune and microbiome profiles have been observed in patients with endometriosis. The objective of this pilot study was to investigate differences in the vaginal and gastrointestinal microbiomes and cervicovaginal immune microenvironment in patients with CPP and endometriosis diagnosis compared to those with CPP without endometriosis and no CPP. METHODS Vaginal swabs, rectal swabs, and cervicovaginal lavages (CVL) were collected among individuals undergoing gynecologic laparoscopy. Participants were grouped based on patients seeking care for chronic pain and/or pathology results: CPP and endometriosis (CPP-Endo) (n = 35), CPP without endometriosis (n = 23), or patients without CPP or endometriosis (controls) (n = 15). Sensitivity analyses were performed on CPP with endometriosis location, stage, and co-occurring gynecologic conditions (abnormal uterine bleeding, fibroids). 16S rRNA sequencing was performed to profile the microbiome, and a panel of soluble immune mediators was quantified using a multiplex assay. Statistical analysis was conducted with SAS, R, MicrobiomeAnalyst, MetaboAnalyst, and QIIME 2. RESULTS Significant differences were observed between participants with CPP alone, CPP-Endo, and surgical controls for body mass index, ethnicity, diagnosis of ovarian cysts, and diagnosis of fibroids. In rectal microbiome analysis, both CPP alone and CPP-Endo exhibited lower alpha diversity than controls, and both CPP groups revealed enrichment of irritable bowel syndrome-associated bacteria. CPP-Endo exhibited an increased abundance of vaginal Streptococcus anginosus and rectal Ruminococcus. Patients with CPP and endometrioma (s) demonstrated increased vaginal Streptococcus, Lactobacillus, and Prevotella compared to other endometriosis sites. Further, abnormal uterine bleeding was associated with an increased abundance of bacterial vaginosis-associated bacteria. Immunoproteomic profiles were distinctly clustered by CPP alone and CPP-Endo compared to controls. CPP-Endo was enriched in TNF⍺, MDC, and IL-1⍺. CONCLUSIONS Vaginal and rectal microbiomes were observed to differ between patients with CPP alone and CPP with endometriosis, which may be useful in personalized treatment for individuals with CPP and endometriosis from those with other causes of CPP. Further investigation is warranted in patients with additional co-occurring conditions, such as AUB/fibroids, which add additional complexity to these conditions and reveal the enrichment of distinct pathogenic bacteria in both mucosal sites. This study provides foundational microbiome-immunoproteomic knowledge related to chronic pelvic pain, endometriosis, and co-occurring gynecologic conditions that can help improve the treatment of patients seeking care for pain.
Collapse
Affiliation(s)
- Nicole Jimenez
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
| | - Taylor Norton
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Department of Obstetrics and Gynecology, Banner University Medical Center Phoenix, Phoenix, AZ, USA
| | - Gurbeen Diadala
- Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Building ABC-1, Lab 331E, 425 N. 5 St, Phoenix, AZ, 85004, USA
| | - Emerald Bell
- Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Building ABC-1, Lab 331E, 425 N. 5 St, Phoenix, AZ, 85004, USA
- University of Arizona College of Nursing, Tucson, AZ, USA
| | - Michelle Valenti
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
| | - Leslie V Farland
- UA Cancer Center, University of Arizona, Tucson, AZ, USA
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA
- Department of Obstetrics and Gynecology, College of Medicine Tucson, University of Arizona, Tucson, AZ, USA
| | - Nichole Mahnert
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA
- Department of Obstetrics and Gynecology, Banner University Medical Center Phoenix, Phoenix, AZ, USA
| | - Melissa M Herbst-Kralovetz
- Department of Obstetrics and Gynecology, College of Medicine-Phoenix, University of Arizona, Phoenix, AZ, USA.
- Basic Medical Sciences, College of Medicine-Phoenix, University of Arizona, Building ABC-1, Lab 331E, 425 N. 5 St, Phoenix, AZ, 85004, USA.
- UA Cancer Center, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
48
|
Schweiger R, Lee S, Zhou C, Yang TP, Smith K, Li S, Sanghvi R, Neville M, Mitchell E, Nessa A, Wadge S, Small KS, Campbell PJ, Sudmant PH, Rahbari R, Durbin R. Insights into non-crossover recombination from long-read sperm sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.05.602249. [PMID: 39005338 PMCID: PMC11245106 DOI: 10.1101/2024.07.05.602249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Meiotic recombination is a fundamental process that generates genetic diversity by creating new combinations of existing alleles. Although human crossovers have been studied at the pedigree, population and single-cell level, the more frequent non-crossover events that lead to gene conversion are harder to study, particularly at the individual level. Here we show that single high-fidelity long sequencing reads from sperm can capture both crossovers and non-crossovers, allowing effectively arbitrary sample sizes for analysis from one male. Using fifteen sperm samples from thirteen donors we demonstrate variation between and within donors for the rates of different types of recombination. Intriguingly, we observe a tendency for non-crossover gene conversions to occur upstream of nearby PRDM9 binding sites, whereas crossover locations have a slight downstream bias. We further provide evidence for two distinct non-crossover processes. One gives rise to the vast majority of non-crossovers with mean conversion tract length under 50bp, which we suggest is an outcome of standard PRDM9-induced meiotic recombination. In contrast ~2% of non-crossovers have much longer mean tract length, and potentially originate from the same process as complex events with more than two haplotype switches, which is not associated with PRDM9 binding sites and is also seen in somatic cells.
Collapse
Affiliation(s)
- Regev Schweiger
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, United Kingdom
| | - Sangjin Lee
- Wellcome Sanger Institute, Cancer Ageing and Somatic Mutation, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Chenxi Zhou
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, United Kingdom
| | - Tsun-Po Yang
- Wellcome Sanger Institute, Cancer Ageing and Somatic Mutation, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Katie Smith
- Wellcome Sanger Institute, Cancer Ageing and Somatic Mutation, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Stacy Li
- Department of Integrative Biology, University of California Berkeley, Berkeley, USA
| | - Rashesh Sanghvi
- Wellcome Sanger Institute, Cancer Ageing and Somatic Mutation, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Matthew Neville
- Wellcome Sanger Institute, Cancer Ageing and Somatic Mutation, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Emily Mitchell
- Wellcome Sanger Institute, Cancer Ageing and Somatic Mutation, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Ayrun Nessa
- Kings College London, Department of Twin Research & Genetic Epidemiology, London, United Kingdom
| | - Sam Wadge
- Kings College London, Department of Twin Research & Genetic Epidemiology, London, United Kingdom
| | - Kerrin S Small
- Kings College London, Department of Twin Research & Genetic Epidemiology, London, United Kingdom
| | - Peter J Campbell
- Wellcome Sanger Institute, Cancer Ageing and Somatic Mutation, Hinxton, Cambridge CB10 1SA, United Kingdom
- Wellcome-MRC Cambridge Stem Cell Institute, Cambridge Biomedical Campus, Cambridge, UK
| | - Peter H Sudmant
- Department of Integrative Biology, University of California Berkeley, Berkeley, USA
- Center for Computational Biology, University of California Berkeley, Berkeley, USA
| | - Raheleh Rahbari
- Wellcome Sanger Institute, Cancer Ageing and Somatic Mutation, Hinxton, Cambridge CB10 1SA, United Kingdom
| | - Richard Durbin
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, United Kingdom
- Wellcome Sanger Institute, Cancer Ageing and Somatic Mutation, Hinxton, Cambridge CB10 1SA, United Kingdom
| |
Collapse
|
49
|
Ahting S, Nährlich L, Held I, Henn C, Krill A, Landwehr K, Meister J, Nährig S, Nolde A, Remke K, Ruppel R, Sauer-Heilborn A, Schebek M, Schopper G, Schulte-Hubbert B, Schwarz C, Smaczny C, Wege S, Hentschel J. Every CFTR variant counts - Target-capture based next-generation-sequencing for molecular diagnosis in the German CF Registry. J Cyst Fibros 2024; 23:774-781. [PMID: 37867076 DOI: 10.1016/j.jcf.2023.10.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/01/2023] [Accepted: 10/09/2023] [Indexed: 10/24/2023]
Abstract
BACKGROUND In times of genotype guided therapy options, a total of 3.2 % of people with CF (pwCF) in the German CF Registry[1] only have one or no CFTR-variant detected after genetic analysis. Additionally, genetic data in the Registry can be documented as free text and can therefore be prone to error. In order to allow the greatest possible amount of pwCF access to modern therapies, we conducted a re-evaluation of free text entries and established a custom-whole-CFTR-locus NGS-approach for all pwCF who remained without genetic confirmation afterwards. METHODS To this end, we assembled 731 free text variants of 655 pwCF in the German CF Registry. All variants were evaluated using ClinVar, HGMD and CFTR1/2, corrected in the Registries' database and uploaded to ClinVar. PwCF whose diagnosis remained uncertain as well as additional pwCF or pwCFTR-RD that were assembled through a nationwide call for testing of unclear cases were offered genetic analysis. Samples were analysed using a target-capture based NGS-custom-design-panel covering the entire CFTR-locus. RESULTS Evaluation of free text variants led to the discovery of 43 variants not formerly reported in the context of CF. The Registries' dropdown list was extended by 497 variants and over 500 pwCF were provided with their most up-to-date genotype. Samples of 47 pwCF/pwCFTR-RD were sequenced via NGS with an overall success rate of 61.7 %, resulting in implementation of entire CFTR-genotyping into routine diagnostics. CONCLUSION Entire CFTR-genotyping can greatly increase the genetic diagnostic rate of pwCF/pwCFTR-RD and should be considered after inconspicuous CFTR screening panels in CFTR-diagnostics.
Collapse
Affiliation(s)
- Simone Ahting
- Institute of Human Genetics, University Medical Center Leipzig, Leipzig, Germany.
| | - Lutz Nährlich
- Department of Pediatrics, Justus-Liebig-University Giessen, Giessen, Germany
| | - Inka Held
- Pediatric Practice Friesenweg, Cystic Fibrosis Center Altona, Hamburg, Germany
| | - Constance Henn
- Division of pediatric Pulmonology and Allergology, Hospital for children and adolescents, University Medical Center Leipzig, Leipzig, Germany
| | - Angelika Krill
- Division of Pneumology, University Medical Center Homburg, Homburg/Saar, Germany
| | - Kerstin Landwehr
- Division of Allergology and Pediatric Pneumology, University Children's Hospital Bethel, University Medical Center Ostwestfalen-Lippe, Bielefeld, Germany
| | - Jochen Meister
- Division of Pneumology, Allergology and Psychotherapy, Children's Hospital, Helios Hospital Aue, Aue, Germany
| | - Susanne Nährig
- Cystic Fibrosis Center for Adults, Med. Klinik V, University Hospital LMU, Munich, Germany
| | - Anna Nolde
- Division of Pneumology, II. Department of Medicine and University Transplant Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katharina Remke
- Department for General Paediatrics, Neonatology and Paediatric Cardiology, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Renate Ruppel
- University Children's Hospital, University Medical Center Erlangen, Erlangen, Germany
| | | | - Martin Schebek
- Division of Pediatric Pneumology, Center for Pediatric and Women's Medicine Kassel, Kassel, Germany
| | - Gudrun Schopper
- University Children's Hospital Schwabing, Technical University of Munich, Munich, Germany
| | - Bernhard Schulte-Hubbert
- Department of medical clinic I, Medical Center Carl Gustav Carus, Technical University of Dresden, Dresden, Germany
| | - Carsten Schwarz
- Department Medicine, HMU-Health and Medical University Potsdam and Director CF Center Westbrandenburg, Division Cystic Fibrosis, Clinic Westbrandenburg, Potsdam, Germany
| | - Christina Smaczny
- Christiane Herzog CF-centre Frankfurt/Main, University Medical Center Frankfurt, Goethe-University Frankfurt, Frankfurt/Main, Germany
| | - Sabine Wege
- Cystic Fibrosis Center, Thoraxklinik Heidelberg, University Medical Center Heidelberg, Heidelberg, Germany
| | - Julia Hentschel
- Institute of Human Genetics, University Medical Center Leipzig, Leipzig, Germany
| |
Collapse
|
50
|
Mishra R, Thunuguntla P, Perkin A, Duraiyan D, Bagwill K, Gonzales S, Brizuela V, Daly S, Chang YJ, Abebe M, Rajana Y, Wichmann K, Bolick C, King J, Fiala M, Fortier J, Jayasinghe R, Schroeder M, Ding L, Vij R, Silva-Fisher J. LINC01432 binds to CELF2 in newly diagnosed multiple myeloma promoting short progression-free survival to standard therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600975. [PMID: 38979159 PMCID: PMC11230414 DOI: 10.1101/2024.06.27.600975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Multiple Myeloma (MM) is a highly prevalent and incurable form of cancer that arises from malignant plasma cells, with over 35,000 new cases diagnosed annually in the United States. While there are a growing number of approved therapies, MM remains incurable and nearly all patients will relapse and exhaust all available treatment options. Mechanisms for disease progression are unclear and in particular, little is known regarding the role of long non-coding RNAs (lncRNA) in mediating disease progression and response to treatment. In this study, we used transcriptome sequencing to compare newly diagnosed MM patients who had short progression-free survival (PFS) to standard first-line treatment (PFS < 24 months) to patients who had prolonged PFS (PFS > 24 months). We identified 157 differentially upregulated lncRNAs with short PFS and focused our efforts on characterizing the most upregulated lncRNA, LINC01432. We investigated LINC01432 overexpression and CRISPR/Cas9 knockdown in MM cell lines to show that LINC01432 overexpression significantly increases cell viability and reduces apoptosis, while knockdown significantly reduces viability and increases apoptosis, supporting the clinical relevance of this lncRNA. Next, we used individual-nucleotide resolution cross-linking immunoprecipitation with RT-qPCR to show that LINC01432 directly interacts with the RNA binding protein, CELF2. Lastly, we showed that LINC01432-targeted locked nucleic acid antisense oligonucleotides reduce viability and increases apoptosis. In summary, this fundamental study identified lncRNAs associated with short PFS to standard NDMM treatment and further characterized LINC01432, which inhibits apoptosis.
Collapse
|