1
|
Leeke BJ, Varsally W, Ogushi S, Zohren J, Menchero S, Courtois A, Snell DM, Teissandier A, Ojarikre O, Mahadevaiah SK, Decarpentrie F, Oakey RJ, VandeBerg JL, Turner JMA. Divergent DNA methylation dynamics in marsupial and eutherian embryos. Nature 2025:10.1038/s41586-025-08992-2. [PMID: 40369084 DOI: 10.1038/s41586-025-08992-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 04/08/2025] [Indexed: 05/16/2025]
Abstract
Based on seminal work in placental species (eutherians)1-10, a paradigm of mammalian development has emerged wherein the genome-wide erasure of parental DNA methylation is required for embryogenesis. Whether such DNA methylation reprogramming is, in fact, conserved in other mammals is unknown. Here, to resolve this point, we generated base-resolution DNA methylation maps in gametes, embryos and adult tissues of a marsupial, the opossum Monodelphis domestica, revealing variations from the eutherian-derived model. The difference in DNA methylation level between oocytes and sperm is less pronounced than that in eutherians. Furthermore, unlike the genome of eutherians, that of the opossum remains hypermethylated during the cleavage stages. In the blastocyst, DNA demethylation is transient and modest in the epiblast. However, it is sustained in the trophectoderm, suggesting an evolutionarily conserved function for DNA hypomethylation in the mammalian placenta. Furthermore, unlike that in eutherians, the inactive X chromosome becomes globally DNA hypomethylated during embryogenesis. We identify gamete differentially methylated regions that exhibit distinct fates in the embryo, with some transient, and others retained and that represent candidate imprinted loci. We also reveal a possible mechanism for imprinted X inactivation, through maternal DNA methylation of the Xist-like noncoding RNA RSX11. We conclude that the evolutionarily divergent eutherians and marsupials use DNA demethylation differently during embryogenesis.
Collapse
Affiliation(s)
- Bryony J Leeke
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK.
- MRC Laboratory of Medical Sciences, London, UK.
- Institute of Clinical Sciences, Imperial College London, London, UK.
| | - Wazeer Varsally
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Sugako Ogushi
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Jasmin Zohren
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Sergio Menchero
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Aurélien Courtois
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Daniel M Snell
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
- Advanced Sequencing Facility, The Francis Crick Institute, London, UK
| | - Aurélie Teissandier
- INSERM U934, CNRS UMR3215, Institut Curie, PSL Research University, Paris, France
| | - Obah Ojarikre
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | | | | | - Rebecca J Oakey
- Department of Medical and Molecular Genetics, King's College London, London, UK
| | - John L VandeBerg
- Division of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - James M A Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
2
|
Wegert J, Appenzeller S, Treger TD, Streitenberger H, Ziegler B, Bausenwein S, Vokuhl C, Parks C, Jüttner E, Gramlich S, Ernestus K, Warman SW, Fuchs J, Hubertus J, von Schweinitz D, Fröhlich B, Jorch N, Knöfler R, Friedrich C, Corbacioglu S, Frühwald MC, Pekrun A, Schneider DT, Faber J, Stursberg J, Metzler M, Welter N, Pritchard-Jones K, Graf N, Furtwängler R, Behjati S, Gessler M. Distinct pathways for genetic and epigenetic predisposition in familial and bilateral Wilms tumor. Genome Med 2025; 17:49. [PMID: 40340749 PMCID: PMC12060375 DOI: 10.1186/s13073-025-01482-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Genetic predisposition is particularly common in children with the kidney cancer, Wilms tumor. In 10% of these children, this manifests as a family history of Wilms tumor or bilateral disease. The frequency and spectrum of underlying changes have not been systematically investigated. METHODS We analyzed 129 children with suspected Wilms tumor predisposition, 20 familial cases, and 109 children with bilateral disease, enrolled over 30 years in the German SIOP93-01/GPOH and SIOP2001 studies. We used whole exome, whole genome, and targeted DNA sequencing, together with MLPA and targeted methylation assays on tumor, blood, and normal kidney to determine predisposing changes. RESULTS Predisposing variants were identified in 117/129 children, comprising DNA variants (57%) and epigenetic changes (34%). Most children had predisposition variants in genes previously implicated in Wilms tumor: most prominently WT1 (n = 35) and less frequently TRIM28, REST, DIS3L2, CTR9, DICER1, CDC73, and NONO. Nine children carried germline mutations in cancer predisposition genes not considered Wilms tumor predisposition genes, such as CHEK2, CDKN2A, BLM, BRCA2, STK11, and FMN2. Predisposition via epigenetic BWS-IC1 alterations occurred as early somatic events, reflected by partial (mosaic) loss of imprinting or loss of heterozygosity at the IGF2/H19 locus in normal kidney or blood. These patients rarely had a clinical diagnosis of Beckwith-Wiedemann syndrome (BWS). Especially WT1-driven tumors follow a stereotypical pathway of germline WT1 mutations becoming homozygous in renal precursor lesions through 11p LOH, which concomitantly activates imprinted IGF2 expression, with subsequent WNT pathway activation leading to tumor growth. There is a high rate of multicentric tumors, which may have previously been missed in unilateral tumors. While Wilms tumor predisposition genes relied on somatic inactivation of the second allele, this was different for general cancer predisposition genes. The latter cases were often associated with additional oncogenic alterations, similar to tumors with epigenetic predisposition. CONCLUSIONS We identified two main mechanisms of Wilms tumor predisposition: either germline genetic alterations of Wilms tumor and, less frequently, general cancer genes; or postzygotic mosaic imprinting defects activating IGF2. These findings inform future genetic screening and risk assessment of affected children and lend support to liquid biopsy screening for enhanced therapeutic stratification.
Collapse
Affiliation(s)
- Jenny Wegert
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Silke Appenzeller
- Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany
| | - Taryn D Treger
- Wellcome Sanger Institute, Hinxton, UK
- Department of Pediatrics, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Heike Streitenberger
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Barbara Ziegler
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Sabrina Bausenwein
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany
| | - Christian Vokuhl
- Section of Pediatric Pathology, Department of Pathology, University Hospital Bonn, Bonn, Germany
| | | | - Eva Jüttner
- Department of Pathology, Schleswig-Holstein University Hospital, Kiel, Germany
| | - Susanne Gramlich
- Department of Pathology, University of Würzburg, Würzburg, Germany
| | - Karen Ernestus
- Department of Pathology, University of Würzburg, Würzburg, Germany
| | - Steven W Warman
- Clinic of Pediatric Surgery, Charité - University Hospital Berlin, Berlin, Germany
| | - Jörg Fuchs
- Department of Pediatric Surgery and Pediatric Urology, University Children's Hospital, Tuebingen, Germany
| | - Jochen Hubertus
- Department of Pediatric Surgery, Marien Hospital Witten, Ruhr-University Bochum, Bochum, Germany
| | - Dietrich von Schweinitz
- Department of Pediatric Surgery, Dr. von Hauner Children's Hospital, LMU University Hospital, Munich, Germany
| | - Birgit Fröhlich
- Department of Pediatric Oncology and Hematology, University of Münster, Münster, Germany
| | - Norbert Jorch
- Evangelisches Klinikum Bethel, Universitätsklinikum OWL, Bielefeld, Germany
| | - Ralf Knöfler
- Department of Pediatric Hematology/Oncology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Carsten Friedrich
- Department of Pediatrics and Pediatric Hematology/Oncology, University Children's Hospital, Carl von Ossietzky University, Klinikum Oldenburg, Oldenburg, Germany
| | - Selim Corbacioglu
- Children's Hospital Regensburg, University of Regensburg, Regensburg, Germany
| | - Michael C Frühwald
- Swabian Children's Cancer Center, Pediatrics and Adolescent Medicine, University Hospital Augsburg, Augsburg, Germany
| | - Arnulf Pekrun
- Pediatric Hematology and Oncology, Klinikum Bremen, Bremen, Germany
| | - Dominik T Schneider
- Clinic of Pediatrics, University Witten/Herdecke, Klinikum Dortmund, Witten, Germany
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center, Johannes Gutenberg-University, Mainz, Germany
| | - Jana Stursberg
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | - Markus Metzler
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Nils Welter
- Department of Pediatric Hematology and Oncology, Saarland University Hospital, Homburg, Germany
| | - Kathy Pritchard-Jones
- UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Norbert Graf
- Department of Pediatric Hematology and Oncology, Saarland University Hospital, Homburg, Germany
| | - Rhoikos Furtwängler
- Department of Pediatric Hematology and Oncology, Saarland University Hospital, Homburg, Germany
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Inselspital University Hospital, University of Bern, Bern, Switzerland
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK
- Department of Pediatrics, University of Cambridge, Cambridge, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Manfred Gessler
- Developmental Biochemistry, Theodor-Boveri-Institute/Biocenter, Julius-Maximilians-University Würzburg, Am Hubland, Würzburg, 97074, Germany.
- Comprehensive Cancer Center Mainfranken, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
3
|
Ware-Gilmore F, Jones MJ, Mejia AJ, Dennington NL, Audsley MD, Hall MD, Sgrò CM, Buckley T, Anand GS, Jose J, McGraw EA. Evolution and adaptation of dengue virus in response to high-temperature passaging in mosquito cells. Virus Evol 2025; 11:veaf016. [PMID: 40330315 PMCID: PMC12054504 DOI: 10.1093/ve/veaf016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/14/2025] [Accepted: 04/23/2025] [Indexed: 05/08/2025] Open
Abstract
The incidence of arboviral diseases like dengue, chikungunya, and yellow fever continues to rise in association with the expanding geographic ranges of their vectors, Aedes aegypti and Aedes albopictus. The distribution of these vectors is believed to be driven in part by climate change and increasing urbanization. Arboviruses navigate a wide range of temperatures as they transition from ectothermic vectors (from 15°C to 35°C) to humans (37°C) and back again, but the role that temperature plays in driving the evolution of arboviruses remains largely unknown. Here, we passaged replicate dengue serotype-2 virus populations 10 times at either 26°C (Low) or 37°C (High) in C6/36 Aedes albopictus cells to explore the differences in adaptation to these thermal environments. We then deep-sequenced the resulting passaged dengue virus populations and tested their replicative fitness in an all-cross temperature regime. We also assessed the ability of the passaged viruses to replicate in the insect vector. While viruses from both thermal regimes accumulated substitutions, only those reared in the 37°C treatments exhibited nonsynonymous changes, including several in the E, or envelope protein, and multiple non-structural genes. Passaging at the higher temperature also led to reduced replicative ability at 26°C in both cells and mosquitoes. One of the mutations in the E gene involved the loss of a glycosylation site previously shown to reduce infectivity in the vector. These findings suggest that viruses selected for growth at higher ambient temperatures may experience tradeoffs between thermostability and replication in the vector. Such associations might also have implications for the suitability of virus transmission under a changing climate.
Collapse
Affiliation(s)
- Fhallon Ware-Gilmore
- Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA
- The Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Matthew J Jones
- Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Austin J Mejia
- Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA
- The Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA
| | - Nina L Dennington
- Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Michelle D Audsley
- School of Biological Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Matthew D Hall
- School of Biological Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Carla M Sgrò
- School of Biological Sciences, Monash University, Melbourne, VIC 3800, Australia
| | - Theresa Buckley
- The Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Ganesh S Anand
- The Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Chemistry, The Pennsylvania State University, University Park, PA 16802, USA
| | - Joyce Jose
- The Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Elizabeth A McGraw
- Department of Entomology, The Pennsylvania State University, University Park, PA 16802, USA
- Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
4
|
Bayó C, Castellano G, Marín F, Castillo-Iturra J, Ocaña T, Kumari H, Pellisé M, Moreira L, Rivero L, Daca-Alvarez M, Ortiz O, Carballal S, Moreira R, Canet-Hermida J, Pineda M, Gabriel C, Flórez-Grau G, Juan M, Benitez-Ribas D, Balaguer F. Discovery and validation of frameshift-derived neopeptides in Lynch syndrome: paving the way for novel cancer prevention strategies. J Immunother Cancer 2025; 13:e011177. [PMID: 40254392 PMCID: PMC12010338 DOI: 10.1136/jitc-2024-011177] [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: 12/05/2024] [Accepted: 03/23/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Lynch syndrome (LS), caused by germline pathogenic variants in the mismatch repair genes, leads to high rates of frameshift-derived neopeptide (FSDN) expression due to microsatellite instability (MSI). While colorectal cancer (CRC) prevention is effective, most LS-related tumors lack such strategies. Cancer vaccines targeting FSDNs offer a promising approach for immune interception in LS. This study aimed to identify and validate LS-related FSDNs to develop vaccines for cancer prevention. METHODS We identified LS-related coding MS mutations and predicted FSDN with high coverage on common Human Leukocyte Antigen (HLA)-I and II alleles. We validated FSDN-associated mutations in colorectal adenomas (CrAD), endometrial cancers (EC), and CRC samples from patients with LS, non-LS tumors, and cell lines. Immunogenicity was assessed through interferon (IFN)-γ enzyme-linked immunospot and flow cytometry analysis of tissue-infiltrating lymphocytes (TILs) from LS carriers. RESULTS We prioritized 53 HLA-I and 45 HLA-II FSDNs in MSI tumors using in silico predictions. Validation revealed 86.7% of FSDN-associated mutations present in LS-CRC samples, with a median of 7.67 (6.5-9) mutations in CrADs and 6.02 (2-10) in CRCs. Sequencing of CrAD and EC samples showed 95% and 77.5% of predicted FSDN-associated mutations, respectively. MSI cancer cell lines transcribed 69.8% of FSDNs. Immunogenicity assays showed that 71% of potential FSDNs elicited IFN-γ responses, with a median of 7.37 (1-10.75) HLA-I and 6 (2-5.75) HLA-II FSDNs per patient. After prioritizing 24 FSDN, in a cohort of 19 LS-derived samples (4 CrAD and 15 normal mucosa), 52% (10/19) demonstrated T-cell reactivity to an HLA-I neoantigen pool. CD8+CD137+ activation markers increased significantly (p=0.037) over time and peptide-specific cells were detected by pentamer staining. CONCLUSIONS Our predicted FSDN set has optimal coverage among LS carriers and can induce IFN-γ inflammatory responses in LS-derived TILs, offering an opportunity for vaccine development.
Collapse
Affiliation(s)
- Cristina Bayó
- Immunology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Giancarlo Castellano
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Fátima Marín
- Hereditary Cancer Program, Catalan institute of oncology, IDIBELL, Badalona, Catalunya, Spain
- Consortium for Biomedical Research in Cancer, Carlos III Institute of Health, CIBERONC, Madrid, Comunidad de Madrid, Spain
| | - Joaquín Castillo-Iturra
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Teresa Ocaña
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Hardeep Kumari
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Maria Pellisé
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Leticia Moreira
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Liseth Rivero
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Maria Daca-Alvarez
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Oswaldo Ortiz
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Sabela Carballal
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Rebeca Moreira
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Julia Canet-Hermida
- Hereditary Cancer Program, Catalan institute of oncology, IDIBELL, Badalona, Catalunya, Spain
- Consortium for Biomedical Research in Cancer, Carlos III Institute of Health, CIBERONC, Madrid, Comunidad de Madrid, Spain
| | - Marta Pineda
- Hereditary Cancer Program, Catalan institute of oncology, IDIBELL, Badalona, Catalunya, Spain
- Consortium for Biomedical Research in Cancer, Carlos III Institute of Health, CIBERONC, Madrid, Comunidad de Madrid, Spain
| | - Capella Gabriel
- Hereditary Cancer Program, Catalan institute of oncology, IDIBELL, Badalona, Catalunya, Spain
- Consortium for Biomedical Research in Cancer, Carlos III Institute of Health, CIBERONC, Madrid, Comunidad de Madrid, Spain
| | - Georgina Flórez-Grau
- Immunology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Manel Juan
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Immunology, Servei d'Immunologia. Hospital Clínic de Barcelona, Barcelona, Barcelona, Spain
| | - Daniel Benitez-Ribas
- Immunology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
- Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Francesc Balaguer
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain
| |
Collapse
|
5
|
Porcher L, Vijayraghavan S, Patel Y, Becker S, Blouin T, McCollum J, Mieczkowski PA, Saini N. Multiple DNA repair pathways prevent acetaldehyde-induced mutagenesis in yeast. Genetics 2025; 229:iyae213. [PMID: 39707916 PMCID: PMC12005267 DOI: 10.1093/genetics/iyae213] [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: 10/23/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024] Open
Abstract
Acetaldehyde is the primary metabolite of alcohol and is present in many environmental sources, including tobacco smoke. Acetaldehyde is genotoxic, whereby it can form DNA adducts and lead to mutagenesis. Individuals with defects in acetaldehyde clearance pathways have increased susceptibility to alcohol-associated cancers. Moreover, a mutation signature specific to acetaldehyde exposure is widespread in alcohol- and smoking-associated cancers. However, the pathways that repair acetaldehyde-induced DNA damage and thus prevent mutagenesis are vaguely understood. Here, we used Saccharomyces cerevisiae to delete genes in each of the major DNA repair pathways to identify those that alter acetaldehyde-induced mutagenesis. We observed that loss of functional nucleotide excision repair had the largest effect on acetaldehyde mutagenesis. In addition, base excision repair and DNA protein crosslink repair pathways were involved in modulating acetaldehyde mutagenesis, while mismatch repair, homologous recombination, and postreplication repair are dispensable for acetaldehyde mutagenesis. Acetaldehyde-induced mutations in a nucleotide excision repair-deficient (Δrad1) background were dependent on translesion synthesis and DNA interstrand crosslink repair. Moreover, whole-genome sequencing of the mutated isolates demonstrated an increase in C→A changes coupled with an enrichment of gCn→A changes, which is diagnostic of acetaldehyde exposure in yeast and in human cancers. Finally, downregulation of the leading strand replicative polymerase Pol epsilon, but not the lagging strand polymerase, resulted in increased acetaldehyde mutagenesis, indicating that lesions are likely formed on the leading strand. Our findings demonstrate that multiple DNA repair pathways coordinate to prevent acetaldehyde-induced mutagenesis.
Collapse
Affiliation(s)
- Latarsha Porcher
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Sriram Vijayraghavan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Yashvi Patel
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Samuel Becker
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Thomas Blouin
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - James McCollum
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Piotr A Mieczkowski
- Department of Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie Saini
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
6
|
Zhu G, Xie Y, Li L, Li R, Sun Y, Zhou T, Cun Y. Single-cell RNA sequencing reveals important role of monocytes and macrophages during mucopolysaccharidosis treatment. Sci Rep 2025; 15:12364. [PMID: 40210734 PMCID: PMC11986106 DOI: 10.1038/s41598-025-97330-7] [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: 12/31/2024] [Accepted: 04/03/2025] [Indexed: 04/12/2025] Open
Abstract
Mucopolysaccharidosis (MPS) encompasses a heterogeneous group of lysosomal storage diseases resulting from mutations in genes encoding lysosomal enzymes responsible for the degradation of mucopolysaccharides, also known as glycosaminoglycans (GAGs). Current therapeutic strategies for MPS include hematopoietic stem cell transplantation (HSCT), enzyme replacement therapy (ERT), and symptomatic therapy. This study investigated dynamic changes in MPS type II (MPS-II) through genomic and single-cell sequencing in a patient undergoing ERT. Analysis of peripheral blood mononuclear cells (PBMCs) from one MPS-II patient of 10 year old at different disease stages through scRNA-seq identified various immune cell types, including natural killer (NK) cells, NKT cells, CD4 + and CD8 + T cells, CD14 + and CD16 + monocytes, and B cells. Monocytes and macrophages were significantly reduced during the severe stage of MPS-II but increased during the recovery stage following ERT. Notably, monocyte subtype mono3 was exclusively expressed in the severe stage, while mono1_2, a subtype of mono1, was absent during the severe stage and exhibited distinct biological functions. These findings suggest that monocytes and macrophages play critical roles in the pathogenesis of MPS-II and in the response to ERT. Pseudotime, Gene Ontology, and cell-communication analyses revealed unique functions for the different cellular subtypes. Notably, key molecules mediating cellular interactions during ERT in MPS-II included CXCR3, PF4, APP, and C5AR1 in macrophages, RPS19 in T cells, HLA-DPB1 in B cells, ADRB2 in NK cells, and IL1B, C5AR1, RPS19, and TNFSF13B in monocytes. Overall, integrative analysis delineated the expression dynamics of various cell types and identified mutations in MPS-II, providing a comprehensive atlas of transcriptional programs, cellular characterizations, and genomic variation profiles in MPS-II. This dataset, along with advanced integrative analysis, represents a valuable resource for the discovery of drug targets and the improvement of therapeutic strategies for MPS-II.
Collapse
Affiliation(s)
- Gaohui Zhu
- Department of Endocrinology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yue Xie
- Pediatric Research Institute, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Li Li
- Department of Endocrinology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Rong Li
- Department of Endocrinology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Yihong Sun
- Pediatric Research Institute, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Ting Zhou
- Department of Endocrinology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| | - Yupeng Cun
- Pediatric Research Institute, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Chongqing, China.
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
| |
Collapse
|
7
|
Ezquerra-Aznárez JM, Gašparovič H, Chiner-Oms Á, Lucía A, Blázquez J, Comas I, Korduláková J, Aínsa JA, Ramón-García S. The emergence of resistance to the antiparasitic selamectin in Mycobacterium smegmatis is improbable and contingent on cell wall integrity. Microbiol Spectr 2025; 13:e0233224. [PMID: 40197087 PMCID: PMC12054103 DOI: 10.1128/spectrum.02332-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/18/2025] [Indexed: 04/09/2025] Open
Abstract
Tuberculosis remains the deadliest infectious disease of the 21st century. New antimicrobials are needed to improve treatment outcomes and enable therapy shortening. Drug repurposing is an alternative to the traditional drug discovery process. The avermectins are a family of macrocyclic lactones with anthelmintic activity active against Mycobacterium tuberculosis. However, their mode of action in mycobacteria remains unknown. In this study, we employed traditional mutant isolation approaches using Mycobacterium smegmatis, a non-pathogenic M. tuberculosis surrogate. We were only able to isolate mutants with decreased susceptibility to selamectin using the ∆nucS mutator M. smegmatis strain. This phenotype was caused by mutations in mps1 and mmpL11. Two of these mutants were used for a second experiment in which high-level selamectin-resistant mutants were isolated; however, specific mutations driving the phenotypic change to high-level resistance could not be identified. The susceptibility to selamectin in these mutants was restored to the basal level by subinhibitory concentrations of ethambutol. The selection of ethambutol resistance in a high-level selamectin-resistant mutant also resulted in multiple colonies becoming susceptible to selamectin again. These colonies carried mutations in embB, suggesting that the integrity of the cell envelope is a prerequisite for selamectin resistance. The absence of increased susceptibility to selamectin in an embB deletion strain demonstrated that the target of selamectin is not cytosolic. Our data show that the concurrence of specific multiple mutations and complete integrity of the mycobacterial envelope are necessary for selamectin resistance. Our studies provide first-time insights into the antimycobacterial mode of action of the antiparasitic avermectins.IMPORTANCETuberculosis is the deadliest infectious disease of the 21st century. New antibiotics are needed to improve treatment. However, developing new drugs is costly and lengthy. Drug repurposing is an alternative to the traditional drug discovery process. The avermectins are a family of drugs used to treat parasitic infections that are active against Mycobacterium tuberculosis, the bacterium that causes tuberculosis. However, their mode of action in mycobacteria remains unknown. Understanding how avermectins kill mycobacteria can facilitate its development as an anti-mycobacterial drug, including against M. tuberculosis.In this study, we used Mycobacterium smegmatis, a non-pathogenic M. tuberculosis surrogate model to understand the molecular mechanisms of how selamectin (a drug of the avermectin family selected for this study as a model) acts against mycobacteria. Our data show that the generation of resistance to selamectin is unlikely and that complete integrity of the mycobacterial envelope is necessary for selamectin resistance, providing first-time insights into the antimycobacterial mode of action of the avermectins.
Collapse
Affiliation(s)
- José Manuel Ezquerra-Aznárez
- Department of Microbiology, Pediatrics, Radiology and Public Health, Faculty of Medicine, and BIFI, University of Zaragoza, Zaragoza, Spain
| | - Henrich Gašparovič
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Álvaro Chiner-Oms
- Genomics of (Re)Emerging Pathogens, Genomics and Health Area, FISABIO–Public Health, Valencia, Spain
- Spanish Network for Research on Epidemiology and Public Health (CIBERESP), Carlos III Health Institute, Madrid, Spain
| | - Ainhoa Lucía
- Department of Microbiology, Pediatrics, Radiology and Public Health, Faculty of Medicine, and BIFI, University of Zaragoza, Zaragoza, Spain
| | - Jesús Blázquez
- National Centre of Biotechnology (CNB-CSIC), Madrid, Spain
| | - Iñaki Comas
- Spanish Network for Research on Epidemiology and Public Health (CIBERESP), Carlos III Health Institute, Madrid, Spain
- Tuberculosis Genomics Unit, Institute for Biomedicine of Valencia (IBV-CSIC), Valencia, Spain
| | - Jana Korduláková
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - José A. Aínsa
- Department of Microbiology, Pediatrics, Radiology and Public Health, Faculty of Medicine, and BIFI, University of Zaragoza, Zaragoza, Spain
- Spanish Network for Research on Respiratory Diseases (CIBERES), Carlos III Health Institute, Madrid, Spain
| | - Santiago Ramón-García
- Department of Microbiology, Pediatrics, Radiology and Public Health, Faculty of Medicine, and BIFI, University of Zaragoza, Zaragoza, Spain
- Spanish Network for Research on Respiratory Diseases (CIBERES), Carlos III Health Institute, Madrid, Spain
- Research and Development Agency of Aragon Foundation (Fundación ARAID), Zaragoza, Spain
| |
Collapse
|
8
|
Lowery FJ, Goff SL, Gasmi B, Parkhurst MR, Ratnam NM, Halas HK, Shelton TE, Langhan MM, Bhasin A, Dinerman AJ, Dulemba V, Goldlust IS, Gustafson AM, Hakim AA, Hitscherich KJ, Kenney LM, Levy L, Rault-Wang JG, Bera A, Ray S, Seavey CD, Hoang CD, Hernandez JM, Gartner JJ, Sindiri S, Prickett TD, McIntyre LS, Krishna S, Robbins PF, Klemen ND, Kwong MLM, Yang JC, Rosenberg SA. Neoantigen-specific tumor-infiltrating lymphocytes in gastrointestinal cancers: a phase 2 trial. Nat Med 2025:10.1038/s41591-025-03627-5. [PMID: 40169866 DOI: 10.1038/s41591-025-03627-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 04/03/2025]
Abstract
Adoptive transfer of unselected autologous tumor-infiltrating lymphocytes (TILs) has mediated meaningful clinical responses in patients with metastatic melanoma but not in cancers of gastrointestinal epithelial origin. In an evolving single-arm phase 2 trial design, TILs were derived from and administered to 91 patients with treatment-refractory mismatch repair proficient metastatic gastrointestinal cancers in a schema with lymphodepleting chemotherapy and high-dose interleukin-2 (three cohorts of an ongoing trial). The primary endpoint of this study was the objective response rate as measured using Response Evaluation Criteria in Solid Tumors 1.0; safety was a descriptive secondary endpoint. In the pilot phase, no clinical responses were observed in 18 patients to bulk, unselected TILs; however, when TILs were screened and selected for neoantigen recognition (SEL-TIL), three responses were seen in 39 patients (7.7% (95% confidence interval (CI): 2.7-20.3)). Based on the high levels of programmed cell death protein 1 in the infused TILs, pembrolizumab was added to the regimen (SEL-TIL + P), and eight objective responses were seen in 34 patients (23.5% (95% CI: 12.4-40.0)). All patients experienced transient severe hematologic toxicities from chemotherapy. Seven (10%) patients required critical care support. Exploratory analyses for laboratory and clinical correlates of response were performed for the SEL-TIL and SEL-TIL + P treatment arms. Response was associated with recognition of an increased number of targeted neoantigens and an increased number of administered CD4+ neoantigen-reactive TILs. The current strategy (SEL-TIL + P) exceeded the parameters of the trial design for patients with colorectal cancer, and an expansion phase is accruing. These results could potentially provide a cell-based treatment in a population not traditionally expected to respond to immunotherapy. ClinicalTrials.gov identifier: NCT01174121 .
Collapse
Affiliation(s)
- Frank J Lowery
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Stephanie L Goff
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA.
| | - Billel Gasmi
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Maria R Parkhurst
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Nivedita M Ratnam
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Hyunmi K Halas
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Thomas E Shelton
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Michelle M Langhan
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Aarushi Bhasin
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Aaron J Dinerman
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Victoria Dulemba
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Ian S Goldlust
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Alexandra M Gustafson
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Abraham A Hakim
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Kyle J Hitscherich
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Lisa M Kenney
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Lior Levy
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Juliette G Rault-Wang
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Alakesh Bera
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Satyajit Ray
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Courtney D Seavey
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Chuong D Hoang
- National Cancer Institute, Center for Cancer Research, Thoracic Surgery Branch, Bethesda, MD, USA
| | - Jonathan M Hernandez
- National Cancer Institute, Center for Cancer Research, Surgical Oncology Program, Bethesda, MD, USA
| | - Jared J Gartner
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Sivasish Sindiri
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Todd D Prickett
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Lori S McIntyre
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Sri Krishna
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Paul F Robbins
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Nicholas D Klemen
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Mei Li M Kwong
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - James C Yang
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA
| | - Steven A Rosenberg
- National Cancer Institute, Center for Cancer Research, Surgery Branch, Bethesda, MD, USA.
| |
Collapse
|
9
|
Thanthirige HA, Wimalarathna NA, Wickramasuriya AM. A comprehensive catalog of single nucleotide polymorphisms (SNPs) from the black pepper (Piper nigrum L.) genome. BMC Genomics 2025; 26:256. [PMID: 40098071 PMCID: PMC11912784 DOI: 10.1186/s12864-025-11414-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 02/27/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Single nucleotide polymorphisms (SNPs) have emerged as the marker of choice in breeding and genetics, particularly in non-model organisms such as black pepper (Piper nigrum L.), a globally recognized spice crop. This study presents a comprehensive catalog of SNPs in the black pepper genome using data from 30 samples obtained from RNA sequencing and restriction site-associated DNA sequencing, retrieved from the Sequence Read Archive, and their consequences at the sequence level. RESULTS Three SNP calling and filtering pipelines, namely BCFtools, Genome Analysis Toolkit (GATK)-soft filtering, and GATK-hard filtering, were employed. Results revealed 498,128, 396,003, and 312,153 SNPs respectively identified by these pipelines, with 260,026 SNPs commonly detected across all methods. Analysis of SNP distribution across the 45 scaffolds of the black pepper genome showed varying densities, with pseudo-chromosomes Pn25 (0.86 SNPs/kb), Pn8 (0.74 SNPs/kb), and Pn7 (0.72 SNPs/kb) exhibiting the highest densities. Conversely, scaffolds Pn27 to Pn43 exhibited minimal SNP distribution, except Pn45. Approximately 34.80% of SNPs exhibited stronger genetic linkage (r2 > 0.7). Moreover, SNPs predominately mapped to downstream (≈ 32.54%), upstream (≈ 22.52%), and exonic (≈ 16.20%) regions of genes. Transition substitution accounted for the majority (≈ 57.42%) of identified SNPs, resulting in an average transition-to-transversion ratio of 1.36. Notably, 56.09% of SNPs were non-synonymous, with a significant proportion (≈ 53.59%) being missense mutations. Additionally, 12,491 SNPs with high or moderate impacts were identified, particularly in genes associated with secondary metabolism and alkaloid biosynthesis pathways. Furthermore, the expression of 675 genes was potentially influenced by local (cis-acting) SNPs, while 554 genes were affected by distal (trans-acting) SNPs. CONCLUSION The findings of the present study underscore the utility of identified SNPs and their targets, especially those impacting important pathways, for future genetic investigations and crop improvement efforts in black pepper. The characterization of SNPs in genes related to secondary metabolism and alkaloid biosynthesis highlights their potential for targeted breeding aimed at enhancing the yield, quality, and resilience of this economically important crop in diverse environmental conditions.
Collapse
Affiliation(s)
- Hiruni A Thanthirige
- Department of Plant Sciences, Faculty of Science, University of Colombo, Colombo, Sri Lanka
| | - Nilni A Wimalarathna
- Department of Plant Sciences, Faculty of Science, University of Colombo, Colombo, Sri Lanka
| | | |
Collapse
|
10
|
Kistler L, de Oliveira Freitas F, Gutaker RM, Maezumi SY, Ramos-Madrigal J, Simon MF, Mendoza F JM, Drovetski SV, Loiselle H, de Oliveira EJ, Vieira EA, Carvalho LJCB, Ellis Perez M, Lin AT, Liu HL, Miller R, Przelomska NAS, Ratan A, Wales N, Wann K, Zhang S, García M, Valenzuela D, Rothhammer F, Santoro CM, Domic AI, Capriles JM, Allaby RG. Historic manioc genomes illuminate maintenance of diversity under long-lived clonal cultivation. Science 2025; 387:eadq0018. [PMID: 40048537 DOI: 10.1126/science.adq0018] [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: 04/24/2024] [Accepted: 12/12/2024] [Indexed: 04/23/2025]
Abstract
Manioc-also called cassava and yuca-is among the world's most important crops, originating in South America in the early Holocene. Domestication for its starchy roots involved a near-total shift from sexual to clonal propagation, and almost all manioc worldwide is now grown from stem cuttings. In this work, we analyze 573 new and published genomes, focusing on traditional varieties from the Americas and wild relatives from herbaria, to reveal the effects of this shift to clonality. We observe kinship over large distances, maintenance of high genetic diversity, intergenerational heterozygosity enrichment, and genomic mosaics of identity-by-descent haploblocks that connect all manioc worldwide. Interviews with Indigenous traditional farmers in the Brazilian Cerrado illuminate how traditional management strategies for sustaining, diversifying, and sharing the gene pool have shaped manioc diversity.
Collapse
Affiliation(s)
- Logan Kistler
- Department of Anthropology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | | | | | - S Yoshi Maezumi
- Department of Coevolution of Land Use and Urbanisation, Max Planck Institute of Geoanthropology, Jena, Germany
| | - Jazmín Ramos-Madrigal
- Center for Evolutionary Hologenomics, Globe Institute, University of Copenhagen, Copenhagen, Denmark
| | - Marcelo F Simon
- Embrapa Recursos Genéticos e Biotecnologia, Brasília, DF, Brazil
| | - J Moises Mendoza F
- Herbario del Oriente Boliviano (USZ), Museo de Historia Natural Noel Kempff Mercado/UAGRM, Santa Cruz, Bolivia
| | - Sergei V Drovetski
- Laboratories for Analytical Biology, Smithsonian Institution, National Museum of Natural History, Washington, DC, USA
| | - Hope Loiselle
- Department of Anthropology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
- Department of Anthropology, University of Washington, Seattle, WA, USA
| | | | | | | | - Marina Ellis Perez
- Department of Anthropology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Audrey T Lin
- Department of Anthropology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
- Richard Gilder Graduate School, American Museum of Natural History, New York, NY, USA
| | - Hsiao-Lei Liu
- Department of Anthropology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
| | - Rachel Miller
- Department of Anthropology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
- The McMaster Ancient DNA Center, Department of Biology, McMaster University, Hamilton, ON, Canada
| | - Natalia A S Przelomska
- Department of Anthropology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
- School of the Environment and Life Sciences, University of Portsmouth, Portsmouth, UK
| | - Aakrosh Ratan
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Nathan Wales
- University of York, BioArCh, Environment Building, Wentworth Way, Heslington, York, UK
| | - Kevin Wann
- Department of Anthropology, National Museum of Natural History, Smithsonian Institution, Washington, DC, USA
- Department of Anthropology, Texas A&M University, College Station, TX, USA
| | - Shuya Zhang
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Magdalena García
- Museo Chileno de Arte Precolumbino, Santiago, Región Metropolitana, Chile
- Millennium Nucleus of Applied Historical Ecology for Arid Forests (AFOREST), Santiago, Chile
| | | | | | - Calogero M Santoro
- Millennium Nucleus of Applied Historical Ecology for Arid Forests (AFOREST), Santiago, Chile
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica, Chile
| | - Alejandra I Domic
- Department of Geosciences, The Pennsylvania State University, University Park, PA, USA
- Department of Anthropology, The Pennsylvania State University, University Park, PA, USA
| | - José M Capriles
- Department of Anthropology, The Pennsylvania State University, University Park, PA, USA
| | - Robin G Allaby
- School of Life Sciences, University of Warwick, Coventry, UK
| |
Collapse
|
11
|
Bai HW, Li N, Zhang YX, Luo JQ, Tian RH, Li P, Huang YH, Bai FR, Deng CZ, Zhao FJ, Mo R, Chi N, Zhou YC, Li Z, Yao CC, Zhi EL. Novel biallelic MCMDC2 variants were associated with meiotic arrest and nonobstructive azoospermia. Asian J Androl 2025; 27:268-275. [PMID: 39789727 PMCID: PMC11949460 DOI: 10.4103/aja202495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/08/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Nonobstructive azoospermia (NOA), one of the most severe types of male infertility, etiology often remains unclear in most cases. Therefore, this study aimed to detect four biallelic detrimental variants (0.5%) in the minichromosome maintenance domain containing 2 ( MCMDC2 ) genes in 768 NOA patients by whole-exome sequencing (WES). Hematoxylin and eosin (H&E) demonstrated that MCMDC2 deleterious variants caused meiotic arrest in three patients (c.1360G>T, c.1956G>T, and c.685C>T) and hypospermatogenesis in one patient (c.94G>T), as further confirmed through immunofluorescence (IF) staining. The single-cell RNA sequencing data indicated that MCMDC2 was substantially expressed during spermatogenesis. The variants were confirmed as deleterious and responsible for patient infertility through bioinformatics and in vitro experimental analyses. The results revealed four MCMDC2 variants related to NOA, which contributes to the current perception of the function of MCMDC2 in male fertility and presents new perspectives on the genetic etiology of NOA.
Collapse
Affiliation(s)
- Hao-Wei Bai
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Na Li
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Yu-Xiang Zhang
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Jia-Qiang Luo
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Ru-Hui Tian
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Peng Li
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Yu-Hua Huang
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Fu-Rong Bai
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Cun-Zhong Deng
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Fu-Jun Zhao
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Ren Mo
- Department of Urology, Inner Mongolia People’s Hospital, Inner Mongolia Urological Institute, Hohhot 010017, China
| | - Ning Chi
- Department of Urology, Inner Mongolia People’s Hospital, Inner Mongolia Urological Institute, Hohhot 010017, China
| | - Yu-Chuan Zhou
- International Peace Maternity and Child Health Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai 200000, China
| | - Zheng Li
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Chen-Cheng Yao
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Er-Lei Zhi
- Department of Andrology, The Center for Men’s Health, Urologic Medical Center, Shanghai Key Laboratory of Reproductive Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| |
Collapse
|
12
|
Beaulieu C, Libourel C, Mbadinga Zamar DL, El Mahboubi K, Hoey DJ, Greiff GRL, Keller J, Girou C, San Clemente H, Diop I, Amblard E, Castel B, Théron A, Cauet S, Rodde N, Zachgo S, Halpape W, Meierhenrich A, Laker B, Bräutigam A, Szovenyi P, Cheng S, Tanizawa Y, Aziz S, Leebens-Mack JH, Schmutz J, Webber J, Grimwood J, Jacquet C, Dunand C, Nelson JM, Roux F, Philippe H, Schornack S, Bonhomme M, Delaux PM. The Marchantia polymorpha pangenome reveals ancient mechanisms of plant adaptation to the environment. Nat Genet 2025; 57:729-740. [PMID: 39962240 PMCID: PMC11906373 DOI: 10.1038/s41588-024-02071-4] [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: 10/27/2023] [Accepted: 12/16/2024] [Indexed: 03/15/2025]
Abstract
Plant adaptation to terrestrial life started 450 million years ago and has played a major role in the evolution of life on Earth. The genetic mechanisms allowing this adaptation to a diversity of terrestrial constraints have been mostly studied by focusing on flowering plants. Here, we gathered a collection of 133 accessions of the model bryophyte Marchantia polymorpha and studied its intraspecific diversity using selection signature analyses, a genome-environment association study and a pangenome. We identified adaptive features, such as peroxidases or nucleotide-binding and leucine-rich repeats (NLRs), also observed in flowering plants, likely inherited from the first land plants. The M. polymorpha pangenome also harbors lineage-specific accessory genes absent from seed plants. We conclude that different land plant lineages still share many elements from the genetic toolkit evolved by their most recent common ancestor to adapt to the terrestrial habitat, refined by lineage-specific polymorphisms and gene family evolution.
Collapse
Grants
- ANR-10-LABX-41 Agence Nationale de la Recherche (French National Research Agency)
- ANR-21-CE20-0010-01 Agence Nationale de la Recherche (French National Research Agency)
- 32022006 National Natural Science Foundation of China (National Science Foundation of China)
- CNRS 80|PRIME MicMac, ERC (grant agreement no. 101001675 - ORIGINS), the project Engineering Nitrogen Symbiosis for Africa (ENSA) currently funded through a grant to the University of Cambridge by the Bill and Melinda Gates Foundation (OPP1172165) and the UK Foreign, Commonwealth and Development Office as Engineering Nitrogen Symbiosis for Africa (OPP1172165).
- Deutsche Forschungsgemeinschaft (ZA, 259/9)
- the URPP Evolution in Action of the University of Zurich, grants of the Swiss National Science Foundation (160004, 131726), the EU’s Horizon 2020 Research and Innovation Program (PlantHUB-No. 722338), the Georges and Antoine Claraz Foundation, and the Forschgungskredit of the University of Zurich (FK-20-089).
- ZhuJiang (2019ZT08N628) and the National Natural Science Foundation of China (32022006)
- the Japan Society for the Promotion of Science KAKENHI (JSPS 20K15783)
- The work (proposal: Award DOI 10.46936/10.25585/60001405) conducted by the U.S. Department of Energy Joint Genome Institute (https://ror.org/04xm1d337), a DOE Office of Science User Facility, is supported by the Office of Science of the U.S. Department of Energy operated under Contract No. DE-AC02-05CH11231
- National Science Foundation (NSF 1501826)
Collapse
Affiliation(s)
- Chloé Beaulieu
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
| | - Cyril Libourel
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
- Unité de Recherche Physiologie, Pathologie et Génétique Végétales, INP PURPAN, Université de Toulouse, Toulouse, France
| | | | - Karima El Mahboubi
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
| | - David J Hoey
- Sainsbury Laboratory, University of Cambridge, Cambridge, UK
| | - George R L Greiff
- Sainsbury Laboratory, University of Cambridge, Cambridge, UK
- University of Bristol, Bristol, UK
| | - Jean Keller
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
| | - Camille Girou
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
| | - Helene San Clemente
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
| | - Issa Diop
- Department of Systematic and Evolutionary Botany, University of Zurich, Zurich, Switzerland
- Zurich-Basel Plant Science Center, Zurich, Switzerland
| | - Emilie Amblard
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
| | - Baptiste Castel
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
| | - Anthony Théron
- CNRGV French Plant Genomic Resource Center, INRAE, Castanet-Tolosan, France
| | - Stéphane Cauet
- CNRGV French Plant Genomic Resource Center, INRAE, Castanet-Tolosan, France
| | - Nathalie Rodde
- CNRGV French Plant Genomic Resource Center, INRAE, Castanet-Tolosan, France
| | - Sabine Zachgo
- Division of Botany, School of Biology, Osnabrueck University, Osnabrueck, Germany
| | - Wiebke Halpape
- Computational Biology, Faculty of Biology, Bielefeld University, Bielefeld, Germany
- CeBiTec, Bielefeld University, Bielefeld, Germany
| | - Anja Meierhenrich
- Computational Biology, Faculty of Biology, Bielefeld University, Bielefeld, Germany
- CeBiTec, Bielefeld University, Bielefeld, Germany
| | - Bianca Laker
- Computational Biology, Faculty of Biology, Bielefeld University, Bielefeld, Germany
- CeBiTec, Bielefeld University, Bielefeld, Germany
| | - Andrea Bräutigam
- Computational Biology, Faculty of Biology, Bielefeld University, Bielefeld, Germany
- CeBiTec, Bielefeld University, Bielefeld, Germany
| | - Peter Szovenyi
- Department of Systematic and Evolutionary Botany, University of Zurich, Zurich, Switzerland
- Zurich-Basel Plant Science Center, Zurich, Switzerland
| | - Shifeng Cheng
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yasuhiro Tanizawa
- Department of Informatics, National Institute of Genetics, Mishima, Japan
| | - Simon Aziz
- Station d'Ecologie Théorique et Expérimentale de Moulis, UMR CNRS 5321, Moulis, France
| | | | - Jeremy Schmutz
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
- Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Jenell Webber
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Jane Grimwood
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Christophe Jacquet
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
| | - Christophe Dunand
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France
| | - Jessica M Nelson
- Maastricht Science Programme, Maastricht University, Maastricht, the Netherlands
| | - Fabrice Roux
- Laboratoire des Interactions Plantes-Microbes-Environnement, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, CNRS, Université de Toulouse, Castanet-Tolosan, France
| | - Hervé Philippe
- Station d'Ecologie Théorique et Expérimentale de Moulis, UMR CNRS 5321, Moulis, France
| | | | - Maxime Bonhomme
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France.
| | - Pierre-Marc Delaux
- Laboratoire de Recherche en Sciences Végétales, Université de Toulouse, CNRS, UPS, Toulouse INP, Castanet-Tolosan, France.
| |
Collapse
|
13
|
Lo HY, Goering R, Kocere A, Lo J, Pockalny M, White L, Ramirez H, Martinez A, Jacobson S, Spitale R, Pearson C, Resendiz ME, Mosimann C, Taliaferro JM. Quantification of subcellular RNA localization through direct detection of RNA oxidation. Nucleic Acids Res 2025; 53:gkaf139. [PMID: 40037712 PMCID: PMC11879412 DOI: 10.1093/nar/gkaf139] [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: 01/10/2024] [Revised: 01/24/2025] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
Across cell types and organisms, thousands of RNAs display asymmetric subcellular distributions. Studying this process requires quantifying abundances of specific RNAs at precise subcellular locations. To analyze subcellular transcriptomes, multiple proximity-based techniques have been developed in which RNAs near a localized bait protein are specifically labeled, facilitating their biotinylation and purification. However, these complex methods are often laborious and require expensive enrichment reagents. To streamline the analysis of localized RNA populations, we developed Oxidation-Induced Nucleotide Conversion sequencing (OINC-seq). In OINC-seq, RNAs near a genetically encoded, localized bait protein are specifically oxidized in a photo-controllable manner. These oxidation events are then directly detected and quantified using high-throughput sequencing and our software package, PIGPEN, without the need for biotin-mediated enrichment. We demonstrate that OINC-seq can induce and quantify RNA oxidation with high specificity in a dose- and light-dependent manner. We further show the spatial specificity of OINC-seq by using it to quantify subcellular transcriptomes associated with the cytoplasm, ER, nucleus, and the inner and outer membranes of mitochondria. Finally, using transgenic zebrafish, we demonstrate that OINC-seq allows proximity-mediated RNA labeling in live animals. In sum, OINC-seq together with PIGPEN provide an accessible workflow for analyzing localized RNAs across different biological systems.
Collapse
Affiliation(s)
- Hei-Yong G Lo
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - Raeann Goering
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - Agnese Kocere
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - Joelle Lo
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - Megan C Pockalny
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - Laura K White
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - Haydee Ramirez
- Department of Chemistry, University of Colorado, Denver, CO, 80204,United States
| | - Abraham Martinez
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - Seth Jacobson
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - Robert C Spitale
- Department of Pharmaceutical Sciences, University of California Irvine, CA, 92697, United States
- Department of Chemistry, University of California Irvine, CA, 92697, United States
| | - Chad G Pearson
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - Marino J E Resendiz
- Department of Chemistry, University of Colorado, Denver, CO, 80204,United States
| | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| | - J Matthew Taliaferro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, United States
| |
Collapse
|
14
|
Mancuso FM, Higareda-Almaraz JC, Canal-Noguer P, Bertossi A, Perera-Lluna A, Roehrl MHA, Kruusmaa K. Colorectal Adenoma Subtypes Exhibit Signature Molecular Profiles: Unique Insights into the Microenvironment of Advanced Precancerous Lesions for Early Detection Applications. Cancers (Basel) 2025; 17:654. [PMID: 40002249 PMCID: PMC11852906 DOI: 10.3390/cancers17040654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/24/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Colorectal cancer (CRC) is characterized by the uncontrolled growth of malignant colonic or rectal crypt epithelium. About 85% of CRCs evolve through a stepwise progression from advanced precancerous adenoma lesions. A better understanding of the evolution from adenoma to carcinoma can provide a window of opportunity not only for early detection and therapeutic intervention but potentially also for cancer prevention strategies. Methods: This study investigates the heterogeneous methylation, copy-number alteration (CNA), and mutation signals of histological adenoma subtypes in the context of progression from normal colon to advanced precancerous lesions (APLs) and early-stage CRC. Results: Differential methylation analysis revealed 2321 significantly altered regions among APLs: 137 hypermethylated regions in serrated vs. tubular, 2093 in serrated vs. tubulovillous, and 91 in tubular vs. tubulovillous adenoma subtypes. The most differentiating pathways for serrated adenomas belonged to cAMP signaling and the regulation of pluripotency of stem cells, while regions separating tubular and tubulovillous subtypes were enriched for WNT signaling. CNA events were mostly present in tubular or tubulovillous adenomas, with the most frequent signals being seen in chromosomes 7, 12, 19, and 20. In contrast, early-stage CRC exhibited signals in chromosomes 7, 8, and 20, indicating different processes between APL and early-stage CRC. Mutations reinforce subtype-level differences, showing specific alterations in each subtype. Conclusions: These findings are especially important for developing early detection or cancer prevention tests trying to capture adenoma signatures.
Collapse
Affiliation(s)
| | | | - Pol Canal-Noguer
- Universal Diagnostics S.A., 41013 Seville, Spain; (F.M.M.); (J.C.H.-A.); (P.C.-N.)
| | - Arianna Bertossi
- Research & Development, Universal Diagnostics d.o.o., 1000 Ljubljana, Slovenia;
| | - Alexandre Perera-Lluna
- B2SLab, Institute for Research and Innovation in Health (IRIS), Universitat Politècnica de Catalunya—BarcelonaTech, 08028 Barcelona, Spain;
- Networking Biomedical Research Centre in the Subject Area of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | | | - Kristi Kruusmaa
- Research & Development, Universal Diagnostics d.o.o., 1000 Ljubljana, Slovenia;
| |
Collapse
|
15
|
Lopez L, Lang PLM, Marciniak S, Kistler L, Latorre SM, Haile A, Cerda EV, Gamba D, Xu Y, Woods P, Yifru M, Kerby J, McKay JK, Oakley CG, Ågren J, Wondimu T, Bulafu C, Perry GH, Burbano HA, Lasky JR. Museum genomics reveals temporal genetic stasis and global genetic diversity in Arabidopsis thaliana. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636844. [PMID: 39975324 PMCID: PMC11839143 DOI: 10.1101/2025.02.06.636844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Global patterns of population genetic variation through time offer a window into evolutionary processes that maintain diversity. Over time, lineages may expand or contract their distribution, causing turnover in population genetic composition. At individual loci, migration, drift, environmental change (among other processes) may affect allele frequencies. Museum specimens of widely distributed species offer a unique window into the genetics of understudied populations and changes over time. Here, we sequenced genomes of 130 herbarium specimens and 91 new field collections of Arabidopsis thaliana and combined these with published genomes. We sought a broader view of genomic diversity across the species, and to test if population genomic composition is changing through time. We documented extensive and previously uncharacterized diversity in a range of populations in Africa, populations that are under threat from anthropogenic climate change. Through time, we did not find dramatic changes in genomic composition of populations. Instead, we found a pattern of genetic change every 100 years of the same magnitude seen when comparing Eurasian populations that are 185 km apart, potentially due to a combination of drift and changing selection. We found only mixed signals of polygenic adaptation at phenology and physiology QTL. We did find that genes conserved across eudicots show altered levels of directional allele frequency change, potentially due to variable purifying and background selection. Our study highlights how museum specimens can reveal new dimensions of population diversity and show how wild populations are evolving in recent history.
Collapse
Affiliation(s)
- Lua Lopez
- Department of Biology, Pennsylvania State University, University Park, PA, USA
- Department of Biology, California State University, San Bernardino, San Bernardino, CA, USA
| | - Patricia L. M. Lang
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Stephanie Marciniak
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA
| | | | - Sergio M. Latorre
- Centre for Life’s Origins and Evolution, Department of Genetics, Evolution and Environment, University College London, London, England, UK
| | - Asnake Haile
- Department of Biology, Pennsylvania State University, University Park, PA, USA
- Department of Plant Biology and Biodiversity Management, Addis Ababa University, Addis Ababa, Ethiopia
| | | | - Diana Gamba
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Yuxing Xu
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Patrick Woods
- Department of Soil and Crop Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Mistire Yifru
- Department of Plant Biology and Biodiversity Management, Addis Ababa University, Addis Ababa, Ethiopia
| | - Jeffrey Kerby
- Aarhus Institute of Advanced Studies, Aarhus, Denmark
| | - John K. McKay
- Department of Soil and Crop Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Christopher G. Oakley
- Department of Botany and Plant Pathology, and The Center for Plant Biology, Purdue University, West Lafayette, IN, USA
| | - Jon Ågren
- Department of Ecology and Genetics, Uppsala University, Uppsala, Sweden
| | - Tigist Wondimu
- Department of Plant Biology and Biodiversity Management, Addis Ababa University, Addis Ababa, Ethiopia
| | - Collins Bulafu
- Department of Plant Sciences, Microbiology, and Biotechnology, Makarere University, Kampala, Uganda
| | - George H. Perry
- Department of Biology, Pennsylvania State University, University Park, PA, USA
- Department of Anthropology, Pennsylvania State University, University Park, PA, USA
| | - Hernán A. Burbano
- Centre for Life’s Origins and Evolution, Department of Genetics, Evolution and Environment, University College London, London, England, UK
| | - Jesse R. Lasky
- Department of Biology, Pennsylvania State University, University Park, PA, USA
- PAC Herbarium, Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
16
|
Moraga C, Branco C, Rougemont Q, Jedlička P, Mendoza-Galindo E, Veltsos P, Hanique M, de la Vega RCR, Tannier E, Liu X, Lemaitre C, Fields PD, Cruaud C, Labadie K, Belser C, Briolay J, Santoni S, Cegan R, Linheiro R, Adam G, Filali AE, Mossion V, Boualem A, Tavares R, Chebbi A, Cordaux R, Fruchard C, Prentout D, Velt A, Spataro B, Delmotte S, Weingartner L, Toegelová H, Tulpová Z, Cápal P, Šimková H, Štorchová H, Krüger M, Abeyawardana OAJ, Taylor DR, Olson MS, Sloan DB, Karrenberg S, Delph LF, Charlesworth D, Muyle A, Giraud T, Bendahmane A, Di Genova A, Madoui MA, Hobza R, Marais GAB. The Silene latifolia genome and its giant Y chromosome. Science 2025; 387:630-636. [PMID: 39913565 PMCID: PMC11890086 DOI: 10.1126/science.adj7430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 04/22/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025]
Abstract
In many species with sex chromosomes, the Y is a tiny chromosome. However, the dioecious plant Silene latifolia has a giant ~550-megabase Y chromosome, which has remained unsequenced so far. We used a long- and short-read hybrid approach to obtain a high-quality male genome. Comparative analysis of the sex chromosomes with their homologs in outgroups showed that the Y is highly rearranged and degenerated. Recombination suppression between X and Y extended in several steps and triggered a massive accumulation of repeats on the Y as well as in the nonrecombining pericentromeric region of the X, leading to giant sex chromosomes. Using sex phenotype mutants, we identified candidate sex-determining genes on the Y in locations consistent with their favoring recombination suppression events 11 and 5 million years ago.
Collapse
Affiliation(s)
- Carol Moraga
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
- Instituto de Ciencias de la Ingeniería, Universidad de O’Higgins, Rancagua, Chile
- Centro UOH de Bioingenieria (CUBI), Universidad de O’Higgins, Rancagua, Chile
| | - Catarina Branco
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO, Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
| | - Quentin Rougemont
- Université Paris-Saclay, CNRS, AgroParisTech, Laboratoire Ecologie Systématique et Evolution, UMR 8079, Bâtiment 680, Gif-sur-Yvette, France
| | - Pavel Jedlička
- Department of Plant Developmental Genetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Eddy Mendoza-Galindo
- Centre d’Ecologie Fonctionnelle et Evolutive (CEFE), University of Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - Paris Veltsos
- Ecology, Evolution and Genetics Research Group, Biology Department, Vrije Universiteit Brussel, Brussels, Belgium
| | - Melissa Hanique
- Institute of Plant Sciences Paris-Saclay (IPS2), Université Paris-Saclay, CNRS, INRAE, Université d’Évry, Gif-sur-Yvette, France
| | - Ricardo C. Rodríguez de la Vega
- Université Paris-Saclay, CNRS, AgroParisTech, Laboratoire Ecologie Systématique et Evolution, UMR 8079, Bâtiment 680, Gif-sur-Yvette, France
| | - Eric Tannier
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
- Inria Lyon Research Center, Villeurbanne, France
| | - Xiaodong Liu
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Claire Lemaitre
- Institut de Recherche en Informatique et Systèmes Aléatoires (IRISA), Université de Rennes, Inria, CNRS, Rennes, France
| | - Peter D. Fields
- Department of Biology, Colorado State University, Fort Collins, CO, USA
| | - Corinne Cruaud
- Genoscope, Institut François Jacob, CEA, CNRS, Université d’Évry, Université Paris-Saclay, Évry, France
| | - Karine Labadie
- Genoscope, Institut François Jacob, CEA, CNRS, Université d’Évry, Université Paris-Saclay, Évry, France
| | - Caroline Belser
- Genoscope, Institut François Jacob, CEA, CNRS, Université d’Évry, Université Paris-Saclay, Évry, France
| | - Jerome Briolay
- Développement de Techniques et Analyse Moléculaire de la Biodiversité (DTAMB), Université Claude Bernard Lyon 1, Campus de la Doua, Villeurbanne, France
| | - Sylvain Santoni
- Genomic Platform, Amélioration Génétique et Adaptation des Plantes Méditerranéennes et Tropicales (AGAP), Université de Montpellier, CIRAD, INRAE, Montpellier, France
| | - Radim Cegan
- Department of Plant Developmental Genetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Raquel Linheiro
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO, Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
| | - Gabriele Adam
- Institute of Plant Sciences Paris-Saclay (IPS2), Université Paris-Saclay, CNRS, INRAE, Université d’Évry, Gif-sur-Yvette, France
| | - Adil El Filali
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Vinciane Mossion
- Department of Ecology and Genetics, Division of Plant Ecology and Evolution, Uppsala University, Uppsala, Sweden
| | - Adnane Boualem
- Institute of Plant Sciences Paris-Saclay (IPS2), Université Paris-Saclay, CNRS, INRAE, Université d’Évry, Gif-sur-Yvette, France
| | - Raquel Tavares
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO, Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
| | - Amine Chebbi
- Efor, Grosspeter Tower (Spaces), Basel, Switzerland
| | - Richard Cordaux
- Évolution Génomes Comportement Écologie, Université Paris-Saclay, CNRS, IRD, Gif-sur-Yvette, France
| | - Cécile Fruchard
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Djivan Prentout
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Amandine Velt
- Santé de la Vigne et Qualité du Vin (SVQV), INRAE, Colmar, France
| | - Bruno Spataro
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Stephane Delmotte
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
| | - Laura Weingartner
- University of Louisville School of Medicine, Undergraduate Medical Education, Louisville, KY, USA
| | - Helena Toegelová
- Institute of Experimental Botany of the Czech Academy of Sciences, Olomouc, Czech Republic
| | - Zuzana Tulpová
- Institute of Experimental Botany of the Czech Academy of Sciences, Olomouc, Czech Republic
| | - Petr Cápal
- Institute of Experimental Botany of the Czech Academy of Sciences, Olomouc, Czech Republic
| | - Hana Šimková
- Institute of Experimental Botany of the Czech Academy of Sciences, Olomouc, Czech Republic
| | - Helena Štorchová
- Plant Reproduction Laboratory, Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | - Manuela Krüger
- Plant Reproduction Laboratory, Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | - Oushadee A. J. Abeyawardana
- Plant Reproduction Laboratory, Institute of Experimental Botany, Czech Academy of Sciences, Prague, Czech Republic
| | - Douglas R. Taylor
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Matthew S. Olson
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Daniel B. Sloan
- Department of Biology, Colorado State University, Fort Collins, CO, USA
| | - Sophie Karrenberg
- Department of Ecology and Genetics, Division of Plant Ecology and Evolution, Uppsala University, Uppsala, Sweden
| | - Lynda F. Delph
- Department of Biology, Indiana University, Bloomington, IN, USA
| | - Deborah Charlesworth
- Institute of Ecology and Evolution, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK
| | - Aline Muyle
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
- Centre d’Ecologie Fonctionnelle et Evolutive (CEFE), University of Montpellier, CNRS, EPHE, IRD, Montpellier, France
| | - Tatiana Giraud
- Université Paris-Saclay, CNRS, AgroParisTech, Laboratoire Ecologie Systématique et Evolution, UMR 8079, Bâtiment 680, Gif-sur-Yvette, France
| | - Abdelhafid Bendahmane
- Institute of Plant Sciences Paris-Saclay (IPS2), Université Paris-Saclay, CNRS, INRAE, Université d’Évry, Gif-sur-Yvette, France
| | - Alex Di Genova
- Instituto de Ciencias de la Ingeniería, Universidad de O’Higgins, Rancagua, Chile
- Centro UOH de Bioingenieria (CUBI), Universidad de O’Higgins, Rancagua, Chile
- Center for Mathematical Modeling, UMI-CNRS 2807, Santiago, Chile
| | - Mohammed-Amin Madoui
- Department of Biology, Colorado State University, Fort Collins, CO, USA
- Service d’Etude des Prions et des Infections Atypiques (SEPIA), Institut François Jacob, Commissariat à l’Energie Atomique et aux Energies Alternatives (CEA), Université Paris Saclay, Fontenay-aux-Roses, France
| | - Roman Hobza
- Department of Plant Developmental Genetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Gabriel A. B. Marais
- Laboratoire Biométrie et Biologie Evolutive (LBBE), CNRS/Université Claude Bernard Lyon 1, Villeurbanne, France
- CIBIO, Centro de Investigação em Biodiversidade e Recursos Genéticos, InBIO, Laboratório Associado, Campus de Vairão, Universidade do Porto, Vairão, Portugal
- Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Porto, Portugal
- BIOPOLIS Program in Genomics, Biodiversity and Land Planning, CIBIO, Campus de Vairão, Vairão, Portugal
- GreenUPorto–Sustainable Agrifood Production Research Centre, Departamento de Biologia, Faculdade de Ciências, Universidade do Porto, Vairão, Portugal
| |
Collapse
|
17
|
Spettel K, Bumberger D, Kriz R, Frank S, Loy M, Galazka S, Suchomel M, Lagler H, Makristathis A, Willinger B. In vitro long-term exposure to chlorhexidine or triclosan induces cross-resistance against azoles in Nakaseomyces glabratus. Antimicrob Resist Infect Control 2025; 14:2. [PMID: 39849551 PMCID: PMC11755926 DOI: 10.1186/s13756-024-01511-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Topical antiseptics are crucial for preventing infections and reducing transmission of pathogens. However, commonly used antiseptic agents have been reported to cause cross-resistance to other antimicrobials in bacteria, which has not yet been described in yeasts. This study aims to assess the in vitro efficacy of antiseptics against clinical and reference isolates of Candida albicans and Nakaseomyces glabratus, and whether prolonged exposure to antiseptics promotes the development of antifungal (cross)resistance. METHODS A high-throughput approach for in vitro resistance development was established to simultaneously expose 96 C. albicans and N. glabratus isolates to increasing concentrations of a given antiseptic - chlorhexidine, triclosan or octenidine. Susceptibility testing and whole genome sequencing of yeast isolates pre- and post-exposure were performed. RESULTS Long-term exposure to antiseptics does not result in the development of stable resistance to the antiseptics themselves. However, 50 N. glabratus isolates acquired resistance to azole antifungals after long-term exposure to triclosan or chlorhexidine, revealing newly acquired mutations in the PDR1 and PMA1 genes. CONCLUSIONS Chlorhexidine as well as triclosan, but not octenidine, were able to introduce selective pressure promoting resistance to azole antifungals. Although we assessed this phenomenon only in vitro, these findings warrant critical monitoring in clinical settings.
Collapse
Affiliation(s)
- Kathrin Spettel
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
- Section Biomedical Science, Health Sciences, FH Campus Wien University of Applied Sciences, Vienna, 1100, Austria
| | - Dominik Bumberger
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Richard Kriz
- Section Biomedical Science, Health Sciences, FH Campus Wien University of Applied Sciences, Vienna, 1100, Austria
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, 1090, Austria
| | - Sarah Frank
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Madita Loy
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Sonia Galazka
- Division of Data, Statistics and Risk Assessment, Austrian Agency for Health and Food Safety AGES, Vienna, 1220, Austria
| | - Miranda Suchomel
- Institute for Hygiene and Applied Immunology, Medical University of Vienna, Vienna, 1090, Austria
| | - Heimo Lagler
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, 1090, Austria
| | - Athanasios Makristathis
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria
| | - Birgit Willinger
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, 1090, Austria.
| |
Collapse
|
18
|
Khan A, Carter R, Mpamhanga CD, Masiga D, Channumsin M, Ciosi M, Manangwa O, Mramba F, Ijaz UZ, Auty H, Mable BK. Swatting Flies: Biting Insects as Non-Invasive Samplers for Mammalian Population Genomics. Mol Ecol 2025:e17661. [PMID: 39838904 DOI: 10.1111/mec.17661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 12/27/2024] [Accepted: 01/10/2025] [Indexed: 01/23/2025]
Abstract
Advances in next-generation sequencing have allowed the use of DNA obtained from unusual sources for wildlife studies. However, these samples have been used predominantly to sequence mitochondrial DNA for species identification while population genetics analyses have been rare. Since next-generation sequencing allows indiscriminate detection of all DNA fragments in a sample, technically it should be possible to sequence whole genomes of animals from environmental samples. Here we used a blood-feeding insect, tsetse fly, to target whole genome sequences of wild animals. Using pools of flies, we compared the ability to recover genomic data from hosts using the short-read sequencing (Illumina) and adaptive sampling of long-read data generated using Oxford nanopore technology (ONT). We found that most of the short-read data (85%-99%) was dominated by tsetse fly DNA and that adaptive sampling on the ONT platform did not substantially reduce this proportion. However, once tsetse reads were removed, the remaining data for both platforms tended to belong to the dominant host expected in the tsetse fly blood meal. Reads mapping to elephants, warthogs and giraffes were recovered more reliably than for buffalo, and there was high variance in the contribution of DNA by individual flies to the pools, suggesting that there are host specific biases. For elephants, using short-read sequencing we were able to identify over 300,000 unfiltered SNPs, which we used to estimate the allele frequencies and expected heterozygosity for the population. Overall, our results show that at least for certain wild mammals, it is possible to recover genome-wide host data from blood-feeding insects.
Collapse
Affiliation(s)
- Anubhab Khan
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
- Department of Biological Sciences, Pwani University, Kilifi, Kenya
- Section for Computational Biology and RNA Biology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Ryan Carter
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Chengetai D Mpamhanga
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Daniel Masiga
- International Centre of Insect Physiology and Ecology (icipe), Nairobi, Kenya
| | - Manun Channumsin
- Faculty of Veterinary Medicine, Rajamangala University of Technology Tawan-Ok, Chonburi, Thailand
| | - Marc Ciosi
- School of Molecular Biosciences, University of Glasgow, Glasgow, UK
| | - Oliva Manangwa
- Vector and Vector-Borne Disease Institute, Tanga, Tanzania
| | | | - Umer Zeeshan Ijaz
- James Watt School of Engineering, University of Glasgow, Glasgow, UK
| | - Harriet Auty
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Barbara K Mable
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
19
|
Lee D, Koo B, Kim S, Byun J, Hong J, Shin DY, Sun CH, Kim J, Song JJ, Jaiswal S, Yoon SS, Kim S, Koh Y. Increased local DNA methylation disorder in AMLs with DNMT3A-destabilizing variants and its clinical implication. Nat Commun 2025; 16:560. [PMID: 39794314 PMCID: PMC11724044 DOI: 10.1038/s41467-024-55691-z] [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: 06/28/2023] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
The mechanistic link between the complex mutational landscape of de novo methyltransferase DNMT3A and the pathology of acute myeloid leukemia (AML) has not been clearly elucidated so far. Motivated by a recent discovery of the significance of DNMT3A-destabilizing mutations (DNMT3AINS) in AML, we here investigate the common characteristics of DNMT3AINS AML methylomes through computational analyses. We present that methylomes of DNMT3AINS AMLs are considerably different from those of DNMT3AR882 AMLs in that they exhibit increased intratumor DNA methylation heterogeneity in bivalent chromatin domains. This epigenetic heterogeneity was associated with the transcriptional variability of developmental and membrane-associated factors shaping stem cell niche, and also was a predictor of the response of AML cells to hypomethylating agents, implying that the survival of AML cells depends on stochastic DNA methylations at bivalent domains. Altogether, our work provides a novel mechanistic model suggesting the genomic origin of the aberrant epigenomic heterogeneity in disease conditions.
Collapse
Affiliation(s)
- Dohoon Lee
- Bioinformatics Institute, Seoul National University, Seoul, Republic of Korea
- BK21 FOUR Intelligence Computing, Seoul National University, Seoul, Republic of Korea
| | - Bonil Koo
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
- AIGENDRUG Co. Ltd, Seoul, Republic of Korea
| | - Seokhyeon Kim
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - Jamin Byun
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Junshik Hong
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dong-Yeop Shin
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea
| | | | - Jaesung Kim
- Department of Biological Sciences, KI for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Ji-Joon Song
- Department of Biological Sciences, KI for BioCentury, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | | | - Sung-Soo Yoon
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.
| | - Sun Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea.
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Seoul, Republic of Korea.
- Department of Computer Science and Engineering, Seoul National University, Seoul, Republic of Korea.
- MOGAM Institute for Biomedical Research, Yong-in, Republic of Korea.
| | - Youngil Koh
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Center for Medical Innovation, Seoul National University Hospital, Seoul, Republic of Korea.
- Genome Opinion Inc, Seoul, Republic of Korea.
| |
Collapse
|
20
|
Maruyama K, Shimizu Y, Nomura Y, Oh-Hara T, Takahashi Y, Nagayama S, Fujita N, Katayama R. Mechanisms of KRAS inhibitor resistance in KRAS-mutant colorectal cancer harboring Her2 amplification and aberrant KRAS localization. NPJ Precis Oncol 2025; 9:4. [PMID: 39762482 PMCID: PMC11704227 DOI: 10.1038/s41698-024-00793-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
KRAS-specific inhibitors have shown promising antitumor effects, especially in non-small cell lung cancer, but limited efficacy in colorectal cancer (CRC) patients. Recent studies have shown that EGFR-mediated adaptive feedback mediates primary resistance to KRAS inhibitors, but the other resistance mechanisms have not been identified. In this study, we investigated intrinsic resistance mechanisms to KRAS inhibitors using patient-derived CRC cells (CRC-PDCs). We found that KRAS-mutated CRC-PDCs can be divided into at least an EGFR pathway-activated group and a PI3K/AKT pathway-activated group. In the latter group, PDCs with PIK3CA major mutation showed high sensitivity to PI3K+mTOR co-inhibition, and a PDC with Her2 amplification with PIK3CA minor mutation showed PI3K-AKT pathway dependency but lost KRAS-MAPK dependency by cytoplasmic localization of KRAS. In the PDC, Her2 knockout restored KRAS plasma membrane localization and KRAS inhibitor sensitivity. The current study provides insight into the mechanisms of primary resistance to KRAS inhibitors, including aberrant KRAS localization.
Collapse
Affiliation(s)
- Kohei Maruyama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuki Shimizu
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yumi Nomura
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama, Japan
- Division of Clinical Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomoko Oh-Hara
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yuki Takahashi
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama, Japan
- Division of Clinical Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Nagayama
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Gastroenterological Surgery, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
- Department of Surgery, Uji-Tokushukai Medical Center, Kyoto, Japan
| | - Naoya Fujita
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan.
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
21
|
Soria E, Lu Q, Boswell W, Du K, Xing Y, Boswell M, Weldon KS, Lai Z, Savage M, Schartl M, Lu Y. Segregation Between an Ornamental and a Disease Driver Gene Provides Insights Into Pigment Cell Regulation. Pigment Cell Melanoma Res 2025; 38:e13196. [PMID: 39289030 DOI: 10.1111/pcmr.13196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/18/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024]
Abstract
Genetic interactions are adaptive within a species. Hybridization can disrupt such species-specific genetic interactions and creates novel interactions that alter the hybrid progeny overall fitness. Hybrid incompatibility, which refers to degenerative genetic interactions that decrease the overall hybrid survival and sterility, is one of the results from combining two diverged genomes in hybrids. The discovery of spontaneous lethal tumorigenesis and underlying genetic interactions in select hybrids between diverged Xiphophorus species showed that lethal pathological process can result from degenerative genetic interactions. Such genetic interactions leading to lethal phenotype are thought to shield gene flow between diverged species. However, hybrids between certain Xiphophorus species do not develop such tumors. Here we report the identification of a locus residing in the genome of one Xiphophorus species that represses an oncogene from a different species. Our finding provides insights into normal and pathological pigment cell development, regulation and a molecular mechanism in hybrid incompatibility.
Collapse
Affiliation(s)
- Erika Soria
- The Xiphophorus Genetic Stock Center, Texas State University, San Marcos, Texas, USA
| | | | - Will Boswell
- The Xiphophorus Genetic Stock Center, Texas State University, San Marcos, Texas, USA
| | - Kang Du
- The Xiphophorus Genetic Stock Center, Texas State University, San Marcos, Texas, USA
| | - Yanting Xing
- The Xiphophorus Genetic Stock Center, Texas State University, San Marcos, Texas, USA
| | - Mikki Boswell
- The Xiphophorus Genetic Stock Center, Texas State University, San Marcos, Texas, USA
| | - Korri S Weldon
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Zhao Lai
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center, San Antonio, Texas, USA
- Department of Molecular Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Markita Savage
- The Xiphophorus Genetic Stock Center, Texas State University, San Marcos, Texas, USA
| | - Manfred Schartl
- The Xiphophorus Genetic Stock Center, Texas State University, San Marcos, Texas, USA
- Developmental Biochemistry, Biocenter, University of Würzburg, Würzburg, Germany
- Research Department for Limnology, University of Innsbruck, Mondsee, Austria
| | - Yuan Lu
- The Xiphophorus Genetic Stock Center, Texas State University, San Marcos, Texas, USA
| |
Collapse
|
22
|
Agostini M, Giacobbi E, Servadei F, Bishof J, Funke L, Sica G, Rovella V, Carilli M, Iacovelli V, Shi Y, Hou J, Candi E, Melino G, Cervelli G, Scimeca M, Mauriello A, Bove P. Unveiling the molecular profile of a prostate carcinoma: implications for personalized medicine. Biol Direct 2024; 19:146. [PMID: 39741346 PMCID: PMC11686862 DOI: 10.1186/s13062-024-00492-z] [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: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Prostate cancer is the most common diagnosed tumor and the fifth cancer related death among men in Europe. Although several genetic alterations such as ERG-TMPRSS2 fusion, MYC amplification, PTEN deletion and mutations in p53 and BRCA2 genes play a key role in the pathogenesis of prostate cancer, specific gene alteration signature that could distinguish indolent from aggressive prostate cancer or may aid in patient stratification for prognosis and/or clinical management of patients with prostate cancer is still missing. Therefore, here, by a multi-omics approach we describe a prostate cancer carrying the fusion of TMPRSS2 with ERG gene and deletion of 16q chromosome arm. RESULTS We have observed deletion of KDM6A gene, which may represent an additional genomic alteration to be considered for patient stratification. The cancer hallmarks gene signatures highlight intriguing molecular aspects that characterize the biology of this tumor by both a high hypoxia and immune infiltration scores. Moreover, our analysis showed a slight increase in the Tumoral Mutational Burden, as well as an over-expression of the immune checkpoints. The omics profiling integrating hypoxia, ROS and the anti-cancer immune response, optimizes therapeutic strategies and advances personalized care for prostate cancer patients. CONCLUSION The here data reported can lay the foundation for predicting a poor prognosis for the studied prostate cancer, as well as the possibility of targeted therapies based on the modulation of hypoxia, ROS, and the anti-cancer immune response.
Collapse
Affiliation(s)
- Massimiliano Agostini
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Erica Giacobbi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Francesca Servadei
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Julia Bishof
- Indivumed GmbH, Falkenried, 88 Building D, 20251, Hamburg, Germany
| | - Likas Funke
- Indivumed GmbH, Falkenried, 88 Building D, 20251, Hamburg, Germany
| | - Giuseppe Sica
- Department of Surgical Science, University Tor Vergata, Viale Oxford 81, 00133, Rome, Italy
| | - Valentina Rovella
- Department of System Medicine, University of Rome "Tor Vergata", 00133, Rome, Italy
| | - Marco Carilli
- Urology Unit, Department of Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Valerio Iacovelli
- Urology Unit, Department of Surgery, Tor Vergata University of Rome, Rome, Italy
| | - Yufang Shi
- Institutes for Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215000, China
| | - Jianquan Hou
- Institutes for Translational Medicine, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215000, China
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Giulio Cervelli
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome "Tor Vergata", Rome, 00133, Italy.
| | - Pierluigi Bove
- Urology Unit, Department of Surgery, Tor Vergata University of Rome, Rome, Italy.
| |
Collapse
|
23
|
Aparicio B, Theunissen P, Hervas-Stubbs S, Fortes P, Sarobe P. Relevance of mutation-derived neoantigens and non-classical antigens for anticancer therapies. Hum Vaccin Immunother 2024; 20:2303799. [PMID: 38346926 PMCID: PMC10863374 DOI: 10.1080/21645515.2024.2303799] [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: 09/29/2023] [Accepted: 01/06/2024] [Indexed: 02/15/2024] Open
Abstract
Efficacy of cancer immunotherapies relies on correct recognition of tumor antigens by lymphocytes, eliciting thus functional responses capable of eliminating tumor cells. Therefore, important efforts have been carried out in antigen identification, with the aim of understanding mechanisms of response to immunotherapy and to design safer and more efficient strategies. In addition to classical tumor-associated antigens identified during the last decades, implementation of next-generation sequencing methodologies is enabling the identification of neoantigens (neoAgs) arising from mutations, leading to the development of new neoAg-directed therapies. Moreover, there are numerous non-classical tumor antigens originated from other sources and identified by new methodologies. Here, we review the relevance of neoAgs in different immunotherapies and the results obtained by applying neoAg-based strategies. In addition, the different types of non-classical tumor antigens and the best approaches for their identification are described. This will help to increase the spectrum of targetable molecules useful in cancer immunotherapies.
Collapse
Affiliation(s)
- Belen Aparicio
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA) University of Navarra, Pamplona, Spain
- Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- CIBERehd, Pamplona, Spain
| | - Patrick Theunissen
- Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- CIBERehd, Pamplona, Spain
- DNA and RNA Medicine Division, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Sandra Hervas-Stubbs
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA) University of Navarra, Pamplona, Spain
- Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- CIBERehd, Pamplona, Spain
| | - Puri Fortes
- Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- CIBERehd, Pamplona, Spain
- DNA and RNA Medicine Division, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Spanish Network for Advanced Therapies (TERAV ISCIII), Spain
| | - Pablo Sarobe
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA) University of Navarra, Pamplona, Spain
- Cancer Center Clinica Universidad de Navarra (CCUN), Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- CIBERehd, Pamplona, Spain
| |
Collapse
|
24
|
Gadji M, Kengne-Ouafo JA, Tchouakui M, Wondji MJ, Mugenzi LMJ, Hearn J, Boyomo O, Wondji CS. Genome-wide association studies unveil major genetic loci driving insecticide resistance in Anopheles funestus in four eco-geographical settings across Cameroon. BMC Genomics 2024; 25:1202. [PMID: 39695386 PMCID: PMC11654272 DOI: 10.1186/s12864-024-11148-7] [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: 06/30/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Insecticide resistance is jeopardising malaria control efforts in Africa. Deciphering the evolutionary dynamics of mosquito populations country-wide is essential for designing effective and sustainable national and subnational tailored strategies to accelerate malaria elimination efforts. Here, we employed genome-wide association studies through pooled template sequencing to compare four eco-geographically different populations of the major vector, Anopheles funestus, across a South North transect in Cameroon, aiming to identify genomic signatures of adaptive responses to insecticides. RESULTS Our analysis revealed limited population structure within Northern and Central regions (FST<0.02), suggesting extensive gene flow, while populations from the Littoral/Coastal region exhibited more distinct genetic patterns (FST>0.049). Greater genetic differentiation was observed at known resistance-associated loci, resistance-to-pyrethroids 1 (rp1) (2R chromosome) and CYP9 (X chromosome), with varying signatures of positive selection across populations. Allelic variation between variants underscores the pervasive impact of selection pressures, with rp1 variants more prevalent in Central and Northern populations (FST>0.3), and the CYP9 associated variants more pronounced in the Littoral/Coastal region (FST =0.29). Evidence of selective sweeps was supported by negative Tajima's D and reduced genetic diversity in all populations, particularly in Central (Elende) and Northern (Tibati) regions. Genomic variant analysis identified novel missense mutations and signatures of complex genomic alterations such as duplications, deletions, transposable element (TE) insertions, and chromosomal inversions, all associated with selective sweeps. A 4.3 kb TE insertion was fixed in all populations with Njombe Littoral/Coastal population, showing higher frequency of CYP9K1 (G454A), a known resistance allele and TE upstream compared to elsewhere. CONCLUSION Our study uncovered regional variations in insecticide resistance candidate variants, emphasizing the need for a streamlined DNA-based diagnostic assay for genomic surveillance across Africa. These findings will contribute to the development of tailored resistance management strategies crucial for addressing the dynamic challenges of malaria control in Cameroon.
Collapse
Affiliation(s)
- Mahamat Gadji
- Centre for Research in Infectious Diseases (CRID), P.O. BOX 13591, Yaoundé, Cameroon.
- The University of Yaoundé 1, P.O BOX 812, Yaoundé, Cameroon.
| | - Jonas A Kengne-Ouafo
- Centre for Research in Infectious Diseases (CRID), P.O. BOX 13591, Yaoundé, Cameroon
| | - Magellan Tchouakui
- Centre for Research in Infectious Diseases (CRID), P.O. BOX 13591, Yaoundé, Cameroon
| | - Murielle J Wondji
- Centre for Research in Infectious Diseases (CRID), P.O. BOX 13591, Yaoundé, Cameroon
- Liverpool School of Tropical Medicine, Pembroke Place Liverpool L3 5QA UK, Liverpool, UK
| | - Leon M J Mugenzi
- Syngenta Crop Protection, Werk Stein, Schaffhauserstrasse, Stein, Switzerland
| | - Jack Hearn
- Centre for Epidemiology and Planetary Health, Scotland's Rural College (SRUC), RAVIC, 9 Inverness Campus, Inverness, UK
| | - Onana Boyomo
- The University of Yaoundé 1, P.O BOX 812, Yaoundé, Cameroon
| | - Charles S Wondji
- Centre for Research in Infectious Diseases (CRID), P.O. BOX 13591, Yaoundé, Cameroon.
- Liverpool School of Tropical Medicine, Pembroke Place Liverpool L3 5QA UK, Liverpool, UK.
| |
Collapse
|
25
|
Li C, Culhane MR, Schroeder DC, Cheeran MCJ, Galina Pantoja L, Jansen ML, Torremorell M. Quantifying the impact of vaccination on transmission and diversity of influenza A variants in pigs. J Virol 2024; 98:e0124524. [PMID: 39530665 DOI: 10.1128/jvi.01245-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Global evolutionary dynamics of influenza A virus (IAV) are fundamentally driven by the extent of virus diversity generated, transmitted, and shaped in individual hosts. How vaccination affects the degree of IAV genetic diversity that can be transmitted and expanded in pigs is unknown. To evaluate the effect of vaccination on the transmission of genetically distinct IAV variants and their diversity after transmission in pigs, we examined the whole genome of IAV recovered from the nasal cavities of pigs vaccinated with different influenza immunization regimens after being infected simultaneously by H1N1 and H3N2 IAVs using a seeder pig model. We found that the seeder pigs harbored more diversified virus populations than the contact pigs. Among contact pigs, H3N2 and H1N1 viruses recovered from pigs vaccinated with a single dose of an unmatched modified live vaccine generally accumulated more extensive genetic mutations than non-vaccinated pigs. Furthermore, the non-sterilizing immunity elicited by the single-dose-modified live vaccine may have exerted positive selection on H1 antigenic regions as we detected significantly higher nonsynonymous but lower synonymous evolutionary rates in H1 antigenic regions than non-antigenic regions. In addition, we observed that the vaccinated pigs shared significantly less proportion of H3N2 variants with seeder pigs than unvaccinated pigs. These results indicated that vaccination might reduce the impact of transmitted influenza variants on the overall diversity of IAV populations harbored in recipient pigs and that within-host genetic selection of IAV is more likely to occur in pigs vaccinated with improperly matched vaccines.IMPORTANCEUnderstanding how vaccination shapes the diversity of influenza variants that transmit and propagate among pigs is essential for designing effective IAV surveillance and control programs. Current knowledge about the transmission of IAV variants has primarily been explored in humans during natural infection. However, how immunity elicited by improperly matched vaccines affects the degree of IAV genetic diversity that can be transmitted and expanded in pigs at the whole-genome level is unknown. We analyzed IAV sequences from samples collected daily from experimentally infected pigs vaccinated with various protocols in a field-represented IAV co-infection model. We found that vaccine-induced non-sterilizing immunity might promote genetic variation on the IAV genome and drive positive selection at antigenic sites during infection. In addition, a smaller proportion of H3N2 viral variants were shared between seeder pigs and vaccinated pigs, suggesting the influence of vaccination on shaping the virus genomic diversity in recipient pigs during the transmission events.
Collapse
Affiliation(s)
- Chong Li
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Marie R Culhane
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Declan C Schroeder
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | - Maxim C-J Cheeran
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA
| | | | | | | |
Collapse
|
26
|
Wang Z, Cao H, Jin J, Thorley E, Cava J, Sun Y, Zhang L, Wang N, Yang Z. Diel asynchrony in the expanded characteristics of toxic cyanobacterial blooms revealed by integrated metabolomics and metagenomics. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136403. [PMID: 39522205 DOI: 10.1016/j.jhazmat.2024.136403] [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: 07/15/2024] [Revised: 10/31/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024]
Abstract
We establish a field metabolomics protocol in Lake Taihu (China) and determined two critical parameters: the minimum amount of biomass for metabolomics and the daytime when metabolomes are stable. The minimum biomass is 475-950 µg dry weight (DW) or 204-408 ng DNA for F (phytoplankton) samples, and 940-1760 µg DW or 193-514 ng DNA for W (whole-water) samples. In a diel cycle, temporal taxonomical composition, metabolic state, and response to physiochemical factors progressed asynchronously between the F and W microbiomes. F peak growth (metabolic steady state) occurred 12-17 pm while W around 12 pm in metabolite identity, concentration, and molecular weight. 482 (∼50 %) metabolites highly correlated between the F and W microbiomes. Integrated analysis revealed different systematic changes between F and W sample, in taxon-associated metabolites, reactions, and biological functions: e.g., carbon metabolism and bioenergetics in F and amino acid metabolism and central metabolism in W samples. Metagenomics discovered important interspecific and intraspecific diversity using single-nucleotide polymorphism, and interactions between cyanobacteria and epibiotic bacteria. Diel intraspecific diversity shift inferred Microcystis aeruginosa and Anabaena sp. have different temperature optima experimentally verified. This integrated multi-omics protocol expands water microbiome analyses from conventional structure and function to diversity dynamics and interspecific metabolism and ecophysiology.
Collapse
Affiliation(s)
- Zeshuang Wang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, School of Biological Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China
| | - Huansheng Cao
- Division of Natural and Applied Sciences, Duke Kunshan University, 8 Duke Ave, Kunshan, Jiangsu 215316, China
| | - Jin Jin
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, School of Biological Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China
| | - Elizabeth Thorley
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, 85287 AZ, USA
| | - John Cava
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, 85287 AZ, USA
| | - Yunfei Sun
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, School of Biological Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China
| | - Lu Zhang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, School of Biological Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China
| | - Nengfei Wang
- School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Zhou Yang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, School of Biological Sciences, Nanjing Normal University, 1 Wenyuan Road, Nanjing 210023, China.
| |
Collapse
|
27
|
Sergio Alberto G, Maximo R, Andres R, Sergio L, Norma P. Comprehensive Analysis of the Influence of Technical and Biological Variations on De Novo Assembly of RNA-Seq Datasets. Bioinform Biol Insights 2024; 18:11779322241274957. [PMID: 39649541 PMCID: PMC11622296 DOI: 10.1177/11779322241274957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 07/25/2024] [Indexed: 12/11/2024] Open
Abstract
De novo assembly of transcriptomes from species without reference genome remains a common problem in functional genomics. While methods and algorithms for transcriptome assembly are continually being developed and published, the quality of de novo assemblies using short reads depends on the complexity of the transcriptome and is limited by several types of errors. One problem to overcome is the research gap regarding the best method to use in each study to obtain high-quality de novo assembly. Currently, there are no established protocols for solving the assembly problem considering the transcriptome complexity. In addition, the accuracy of quality metrics used to evaluate assemblies remains unclear. In this study, we investigate and discuss how different variables accounting for the complexity of RNA-Seq data influence assembly results independently of the software used. For this purpose, we simulated transcriptomic short-read sequence datasets from high-quality full-length predicted transcript models with varying degrees of complexity. Subsequently, we conducted de novo assemblies using different assembly programs, and compared and classified the results using both reference-dependent and independent metrics. These metrics were assessed both individually and combined through multivariate analysis. The degree of alternative splicing and the fragment size of the paired-end reads were identified as the variables with the greatest influence on the assembly results. Moreover, read length and fragment size had different influences on the reconstruction of longer and shorter transcripts. These results underscore the importance of understanding the composition of the transcriptome under study, and making experimental design decisions related to the need to work with reads and fragments of different sizes. In addition, the choice of assembly software will positively impact the final assembly outcome. This selection will affect the completeness of represented genes and assembled isoforms, as well as contribute to error reduction.
Collapse
Affiliation(s)
- Gonzalez Sergio Alberto
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires, Argentina
| | - Rivarola Maximo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ribone Andres
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Lew Sergio
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Instituto de Ingeniería Biomédica, Facultad de Ingeniería, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Paniego Norma
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), CICVyA, Instituto Nacional de Tecnología Agropecuaria (INTA), Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| |
Collapse
|
28
|
Pokorna P, Palova H, Adamcova S, Jugas R, Al Tukmachi D, Kyr M, Knoflickova D, Kozelkova K, Bystry V, Mejstrikova S, Merta T, Trachtova K, Podlipna E, Mudry P, Pavelka Z, Bajciova V, Tinka P, Jarosova M, Catela Ivkovic T, Madlener S, Pal K, Stepien N, Mayr L, Tichy B, Drabova K, Jezova M, Kozakova S, Vanackova J, Radova L, Steininger K, Haberler C, Gojo J, Sterba J, Slaby O. Real-World Performance of Integrative Clinical Genomics in Pediatric Precision Oncology. J Transl Med 2024; 104:102161. [PMID: 39442669 DOI: 10.1016/j.labinv.2024.102161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/16/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
Despite significant improvement in the survival of pediatric patients with cancer, treatment outcomes for high-risk, relapsed, and refractory cancers remain unsatisfactory. Moreover, prolonged survival is frequently associated with long-term adverse effects due to intensive multimodal treatments. Accelerating the progress of pediatric oncology requires both therapeutic advances and strategies to mitigate the long-term cytotoxic side effects, potentially through targeting specific molecular drivers of pediatric malignancies. In this report, we present the results of integrative genomic and transcriptomic profiling of 230 patients with malignant solid tumors (the "primary cohort") and 18 patients with recurrent or otherwise difficult-to-treat nonmalignant conditions (the "secondary cohort"). The integrative workflow for the primary cohort enabled the identification of clinically significant single nucleotide variants, small insertions/deletions, and fusion genes, which were found in 55% and 28% of patients, respectively. For 38% of patients, molecularly informed treatment recommendations were made. In the secondary cohort, known or potentially driving alteration was detected in 89% of cases, including a suspected novel causal gene for patients with inclusion body infantile digital fibromatosis. Furthermore, 47% of findings also brought therapeutic implications for subsequent management. Across both cohorts, changes or refinements to the original histopathological diagnoses were achieved in 4% of cases. Our study demonstrates the efficacy of integrating advanced genomic and transcriptomic analyses to identify therapeutic targets, refine diagnoses, and optimize treatment strategies for challenging pediatric and young adult malignancies and underscores the need for broad implementation of precision oncology in clinical settings.
Collapse
Affiliation(s)
- Petra Pokorna
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic; Center for Precision Medicine, University Hospital Brno, Brno, Czech Republic
| | - Hana Palova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sona Adamcova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Robin Jugas
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dagmar Al Tukmachi
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Biochemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Michal Kyr
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dana Knoflickova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Katerina Kozelkova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Vojtech Bystry
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Sona Mejstrikova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tomas Merta
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Karolina Trachtova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Eliska Podlipna
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Peter Mudry
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zdenek Pavelka
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Viera Bajciova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Tinka
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marie Jarosova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tina Catela Ivkovic
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Sibylle Madlener
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Karol Pal
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Natalia Stepien
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Lisa Mayr
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Boris Tichy
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Klara Drabova
- Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marta Jezova
- Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sarka Kozakova
- Department of Pharmacology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Department of Pharmacy, University Hospital Brno, Brno, Czech Republic
| | - Jitka Vanackova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Lenka Radova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Karin Steininger
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Christine Haberler
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Johannes Gojo
- Department of Pediatrics and Adolescent Medicine, Comprehensive Center for Pediatrics and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Jaroslav Sterba
- Center for Precision Medicine, University Hospital Brno, Brno, Czech Republic; Department of Pediatric Oncology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic; Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic; Center for Precision Medicine, University Hospital Brno, Brno, Czech Republic; Department of Pathology, University Hospital Brno and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
29
|
Vatsyayan A, Imran M, Bhardwaj J, Vr A, Agrawal SJ, Saikia BJ, Senthivel V, Pandhare K, Bhoyar RC, Divakar MK, Mishra A, Jolly B, Trehan S, Sivasubbu S, Scaria V. Understanding the variant landscape, and genetic epidemiology of Multiple Endocrine Neoplasia in India. Endocrine 2024; 86:1178-1187. [PMID: 39112918 DOI: 10.1007/s12020-024-03982-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/24/2024] [Indexed: 11/12/2024]
Abstract
PURPOSE Multiple Endocrine Neoplasia (MEN) is a group of familial cancer syndromes that encompasses several types of endocrine tumors differentiated by genetic mutations in RET, MEN1 and CDKN1B genes. Accurate diagnosis of MEN subtypes can thus be performed through genetic testing. However, MEN variants remain largely understudied in Indian populations. Additionally, few dedicated resources to understand these disorders currently exist. METHODS Using the gold-standard ACMG/AMP guidelines, we systematically classified variants reported across the three genes in the IndiGen dataset, and established the genetic epidemiology of MEN in the Indian population. We further classified ClinVar and Mastermind variants and compiled all into a database. Finally, we designed a multiplex primer panel for rapid variant identification. RESULTS We have established the genetic prevalence of MEN as the following: 1 in 1026 individuals is likely to be afflicted with MEN linked with pathogenic RET mutations. We have further created the MAPVar database containing 3280 ACMG-classified variants freely accessible at: https://clingen.igib.res.in/MAPVar/ . Finally, our NGS primer panel covers 33 exonic regions across two pools through 38 amplicons with a total amplified region of 65 kb. CONCLUSION Our work establishes that MEN is a prevalent disorder in India. The rare nature of Indian variants underscores the need of genomic and functional studies to establish a more comprehensive variant landscape. Additionally, our panel offers a means of cost-effective genetic testing, and the MAPVar database a ready reference to aid in a better understanding of variant pathogenicity in clinical as well as research settings.
Collapse
Affiliation(s)
- Aastha Vatsyayan
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Mohamed Imran
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Juhi Bhardwaj
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Arvinden Vr
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srashti Jyoti Agrawal
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Bhaskar Jyoti Saikia
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Vigneshwar Senthivel
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kavita Pandhare
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rahul C Bhoyar
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
| | - Mohit Kumar Divakar
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Anushree Mishra
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
| | - Bani Jolly
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Suruchi Trehan
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sridhar Sivasubbu
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Vishwanath Cancer Care Foundation, B 702, Neelkanth Business Park Kirol Village, Mumbai, 400086, India
| | - Vinod Scaria
- CSIR Institute of Genomics and Integrative Biology (CSIR-IGIB) Mathura Road, Delhi, 110025, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
- Dr. D. Y. Patil Medical College, Hospital & Research Centre, Pune, India.
- Vishwanath Cancer Care Foundation, B 702, Neelkanth Business Park Kirol Village, Mumbai, 400086, India.
| |
Collapse
|
30
|
Duan H, Ren J, Wei S, Yang Z, Li C, Wang Z, Li M, Wei Z, Liu Y, Wang X, Lan H, Zeng Z, Xie M, Xie Y, Wu S, Hu W, Guo C, Zhang X, Liang L, Yu C, Mou Y, Jiang Y, Li H, Sugarman E, Deek RA, Chen Z, Li T, Chen Y, Yao M, Chen L, Liu L, Zhang G, Mou Y. Integrated analyses of multi-omic data derived from paired primary lung cancer and brain metastasis reveal the metabolic vulnerability as a novel therapeutic target. Genome Med 2024; 16:138. [PMID: 39593114 PMCID: PMC11590298 DOI: 10.1186/s13073-024-01410-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Lung cancer brain metastases (LC-BrMs) are frequently associated with dismal mortality rates in patients with lung cancer; however, standard of care therapies for LC-BrMs are still limited in their efficacy. A deep understanding of molecular mechanisms and tumor microenvironment of LC-BrMs will provide us with new insights into developing novel therapeutics for treating patients with LC-BrMs. METHODS Here, we performed integrated analyses of genomic, transcriptomic, proteomic, metabolomic, and single-cell RNA sequencing data which were derived from a total number of 154 patients with paired and unpaired primary lung cancer and LC-BrM, spanning four published and two newly generated patient cohorts on both bulk and single cell levels. RESULTS We uncovered that LC-BrMs exhibited a significantly greater intra-tumor heterogeneity. We also observed that mutations in a subset of genes were almost always shared by both primary lung cancers and LC-BrM lesions, including TTN, TP53, MUC16, LRP1B, RYR2, and EGFR. In addition, the genome-wide landscape of somatic copy number alterations was similar between primary lung cancers and LC-BrM lesions. Nevertheless, several regions of focal amplification were significantly enriched in LC-BrMs, including 5p15.33 and 20q13.33. Intriguingly, integrated analyses of transcriptomic, proteomic, and metabolomic data revealed mitochondrial-specific metabolism was activated but tumor immune microenvironment was suppressed in LC-BrMs. Subsequently, we validated our results by conducting real-time quantitative reverse transcription PCR experiments, immunohistochemistry, and multiplexed immunofluorescence staining of patients' paired tumor specimens. Therapeutically, targeting oxidative phosphorylation with gamitrinib in patient-derived organoids of LC-BrMs induced apoptosis and inhibited cell proliferation. The combination of gamitrinib plus anti-PD-1 immunotherapy significantly improved survival of mice bearing LC-BrMs. Patients with a higher expression of mitochondrial metabolism genes but a lower expression of immune genes in their LC-BrM lesions tended to have a worse survival outcome. CONCLUSIONS In conclusion, our findings not only provide comprehensive and integrated perspectives of molecular underpinnings of LC-BrMs but also contribute to the development of a potential, rationale-based combinatorial therapeutic strategy with the goal of translating it into clinical trials for patients with LC-BrMs.
Collapse
Affiliation(s)
- Hao Duan
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Jianlan Ren
- Department of Computer Science, Ying Wu College of Computing, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Shiyou Wei
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenyu Yang
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chuan Li
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhenning Wang
- Department of Neurosurgery, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, 523018, China
| | - Meichen Li
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Zhi Wei
- Department of Computer Science, Ying Wu College of Computing, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
| | - Yu Liu
- Faculty of Dentistry, Prince Philip Dental Hospital, the University of Hong Kong, Sai Ying Pun, Hong Kong, China
| | - Xiuqi Wang
- Faculty of Dentistry, Prince Philip Dental Hospital, the University of Hong Kong, Sai Ying Pun, Hong Kong, China
| | - Hongbin Lan
- Faculty of Dentistry, Prince Philip Dental Hospital, the University of Hong Kong, Sai Ying Pun, Hong Kong, China
| | - Zhen Zeng
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Maodi Xie
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuan Xie
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Suwen Wu
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Wanming Hu
- Department of Pathology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Chengcheng Guo
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Xiangheng Zhang
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Lun Liang
- Department of Neurosurgery, The First Affiliated Hospital, Guangxi Medical University, Nanning, 530021, China
| | - Chengwei Yu
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-Sen University Lingnan Hospital, Guangzhou, 510530, China
| | - Yanhao Mou
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yu Jiang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Houde Li
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Eric Sugarman
- Philadelphia College of Osteopathic Medicine, Philadelphia, PA, 19131, USA
| | - Rebecca A Deek
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Zexin Chen
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou, 510535, China
| | - Tao Li
- Department of Anesthesiology, Laboratory of Mitochondrial Metabolism and Perioperative Medicine, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yaohui Chen
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Maojin Yao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, China.
| | - Likun Chen
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Lunxu Liu
- Department of Thoracic Surgery, Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Gao Zhang
- Faculty of Dentistry, Prince Philip Dental Hospital, the University of Hong Kong, Sai Ying Pun, Hong Kong, China.
| | - Yonggao Mou
- Department of Neurosurgery/Neuro-Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| |
Collapse
|
31
|
Legan AW, Allan CW, Jensen ZN, Degain BA, Yang F, Kerns DL, Benowitz KM, Fabrick JA, Li X, Carrière Y, Matzkin LM, Tabashnik BE. Mismatch between lab-generated and field-evolved resistance to transgenic Bt crops in Helicoverpa zea. Proc Natl Acad Sci U S A 2024; 121:e2416091121. [PMID: 39503848 PMCID: PMC11588094 DOI: 10.1073/pnas.2416091121] [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: 08/08/2024] [Accepted: 10/03/2024] [Indexed: 11/27/2024] Open
Abstract
Transgenic crops producing crystalline (Cry) proteins from the bacterium Bacillus thuringiensis (Bt) have been used extensively to control some major crop pests. However, many populations of the noctuid moth Helicoverpa zea, one of the most important crop pests in the United States, have evolved practical resistance to several Cry proteins including Cry1Ac. Although mutations in single genes that confer resistance to Cry proteins have been identified in lab-selected and gene-edited strains of H. zea and other lepidopteran pests, the genetic basis of field-evolved resistance to Cry proteins in H. zea has remained elusive. We used a genomic approach to analyze the genetic basis of field-evolved resistance to Cry1Ac in 937 H. zea derived from 17 sites in seven states of the southern United States. We found evidence for extensive gene flow among all populations studied. Field-evolved resistance was not associated with mutations in 20 single candidate genes previously implicated in resistance or susceptibility to Cry proteins in H. zea or other lepidopterans. Instead, resistance in field samples was associated with increased copy number of a cluster of nine trypsin genes. However, trypsin gene amplification occurred in a susceptible sample and not in all resistant samples, implying that this amplification does not always confer resistance and mutations in other genes also contribute to field-evolved resistance to Cry1Ac in H. zea. The mismatch between lab-generated and field-evolved resistance in H. zea is unlike other cases of Bt resistance and reflects challenges for managing this pest.
Collapse
Affiliation(s)
- Andrew W. Legan
- Department of Entomology, University of Arizona, Tucson, AZ85721
| | - Carson W. Allan
- Department of Entomology, University of Arizona, Tucson, AZ85721
| | - Zoe N. Jensen
- Department of Entomology, University of Arizona, Tucson, AZ85721
| | | | - Fei Yang
- Department of Entomology, University of Minnesota, St. Paul, MN55108
| | - David L. Kerns
- Department of Entomology, Texas A&M University, College Station, TX77843
| | - Kyle M. Benowitz
- College of Integrative Sciences and Arts, Arizona State University, Mesa, AZ85212
| | - Jeffrey A. Fabrick
- US Department of Agriculture, Agricultural Research Service, US Arid Land Agricultural Research Center, Maricopa, AZ85138
| | - Xianchun Li
- Department of Entomology, University of Arizona, Tucson, AZ85721
| | - Yves Carrière
- Department of Entomology, University of Arizona, Tucson, AZ85721
| | | | | |
Collapse
|
32
|
Lo HYG, Goering R, Kocere A, Lo J, Pockalny MC, White LK, Ramirez H, Martinez A, Jacobson S, Spitale RC, Pearson CG, Resendiz MJE, Mosimann C, Taliaferro JM. Quantification of subcellular RNA localization through direct detection of RNA oxidation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.12.623278. [PMID: 39605352 PMCID: PMC11601319 DOI: 10.1101/2024.11.12.623278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Across cell types and organisms, thousands of RNAs display asymmetric subcellular distributions. The study of this process often requires quantifying abundances of specific RNAs at precise subcellular locations. To analyze subcellular transcriptomes, multiple proximity-based techniques have been developed in which RNAs near a localized bait protein are specifically labeled, facilitating their biotinylation and purification. However, these complex methods are often laborious and require expensive enrichment reagents. To streamline the analysis of localized RNA populations, we developed Oxidation-Induced Nucleotide Conversion sequencing (OINC-seq). In OINC-seq, RNAs near a genetically encoded, localized bait protein are specifically oxidized in a photo-controllable manner. These oxidation events are then directly detected and quantified using high-throughput sequencing and our software package, PIGPEN, without the need for biotin-mediated enrichment. We demonstrate that OINC-seq can induce and quantify RNA oxidation with high specificity in a dose- and light-dependent manner. We further show the spatial specificity of OINC-seq by using it to quantify subcellular transcriptomes associated with the cytoplasm, ER, nucleus, and the inner and outer membranes of mitochondria. Finally, using transgenic zebrafish, we demonstrate that OINC-seq allows proximity-mediated RNA labeling in live animals. In sum, OINC-seq together with PIGPEN provide an accessible workflow for the analysis of localized RNAs across different biological systems.
Collapse
Affiliation(s)
- Hei-Yong G. Lo
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, USA
| | - Raeann Goering
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, USA
| | - Agnese Kocere
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, USA
| | - Joelle Lo
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, USA
| | - Megan C. Pockalny
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, USA
| | - Laura K. White
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, USA
| | - Haydee Ramirez
- Department of Chemistry, University of Colorado, Denver, USA
| | - Abraham Martinez
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, USA
| | - Seth Jacobson
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, USA
| | - Robert C. Spitale
- Department of Pharmaceutical Sciences, University of California Irvine, USA
- Department of Chemistry, University of California Irvine, USA
| | - Chad G. Pearson
- Department of Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, USA
| | | | - Christian Mosimann
- Department of Pediatrics, Section of Developmental Biology, University of Colorado Anschutz Medical Campus, USA
| | - J. Matthew Taliaferro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, USA
| |
Collapse
|
33
|
Imran M, Arvinden VR, Mehanathan PB, Rajagopal RE, Muthu SP, Arunachalam AS, Bhoyar RC, Vignesh H, Mitra S, Jha GN, Gupta A, Kumar M, Bhowmick R, Bhunia NS, Dutta AK, Scaria V, Sivasubbu S. A Rapid and Scalable Multiplex PCR-Based Next-Generation Amplicon Sequencing Method for Familial Hypercholesterolemia Genetic Screening. J Appl Lab Med 2024; 9:871-885. [PMID: 39140510 DOI: 10.1093/jalm/jfae089] [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: 03/21/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024]
Abstract
BACKGROUND Familial hypercholesterolemia (FH) is a frequently underdiagnosed genetic disorder characterized by elevated low-density lipoprotein (LDL) levels. Genetic testing of LDLR, APOB, and PCSK9 genes can identify variants in up to 80% of clinically diagnosed patients. However, limitations in time, scalability, and cost have hindered effective next-generation sequencing of these genes. Additionally, pharmacogenomic variants are associated with statin-induced adverse effects in FH patients. To address these challenges, we developed a multiplex primer-based amplicon sequencing approach for FH genetic testing. METHODS Multiplex primers were designed for the exons of the LDLR, APOB, and PCSK9 genes, as well as for pharmacogenomic variants rs4149056 (SLCO1B1:c.521T > A), rs2306283 (SLCO1B1:c.388A > G), and rs2231142 (ABCG2:c.421C > A). Analytical validation using samples with known pathogenic variants and clinical validation with 12 FH-suspected probands were conducted. Library preparation was based on a bead-based tagmentation method, and sequencing was conducted on the NovaSeq 6000 platform. RESULTS Our approach ensured no amplicon dropouts, with over 100× coverage on each amplicon. Known variants in 2 samples were successfully detected. Further, we identified one heterozygous LDLR (p.Glu228Ter) variant and 2 homozygous cases of LDLR (p.Lys294Ter) and LDLR (p.Ser177Leu) variants in patients. Pharmacogenomic analysis revealed that overall 3 patients may require reduced statin doses. Our approach offered reduced library preparation time (approximately 3 h), greater scalability, and lower costs (under $50) for FH genetic testing. CONCLUSIONS Our method effectively sequences LDLR, APOB, and PCSK9 genes including pharmacogenomic variants that will guide appropriate screening and statin dosing, thus increasing both efficiency and affordability.
Collapse
Affiliation(s)
- Mohamed Imran
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - V R Arvinden
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | | | - Raskin Erusan Rajagopal
- Multidisciplinary Research Unit, Tirunelveli Medical College Hospital, Tirunelveli, Tamil Nadu, India
| | - Suriya Prabha Muthu
- Multidisciplinary Research Unit, Tirunelveli Medical College Hospital, Tirunelveli, Tamil Nadu, India
| | | | - Rahul C Bhoyar
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Harie Vignesh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Samya Mitra
- Department of Pediatrics, AIIMS Kalyani, Kalyani West Bengal, India
| | - Ganga Nath Jha
- Department of Anthropology, Vinoba Bhave University, Hazaribag, Jharkhand, India
| | - Aayush Gupta
- Department of Dermatology, Dr. D.Y Patil Medical College, Hospital and Research Centre, Pimpri, Pune, Maharashtra, India
| | - Manoj Kumar
- Department of Zoology, Vinoba Bhave University, Hazaribag Jharkhand, India
| | - Rohit Bhowmick
- Department of Pediatrics, AIIMS Kalyani, Kalyani West Bengal, India
| | | | - Atanu Kumar Dutta
- Department of Biochemistry, AIIMS Kalyani, Kalyani, West Bengal, India
| | - Vinod Scaria
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
34
|
Kriz R, Spettel K, Pichler A, Schefberger K, Sanz-Codina M, Lötsch F, Harrison N, Willinger B, Zeitlinger M, Burgmann H, Lagler H. In vitro resistance development gives insights into molecular resistance mechanisms against cefiderocol. J Antibiot (Tokyo) 2024; 77:757-767. [PMID: 39080477 PMCID: PMC11513634 DOI: 10.1038/s41429-024-00762-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 10/29/2024]
Abstract
Cefiderocol, a novel siderophore cephalosporin, demonstrates promising in vitro activity against multidrug-resistant Gram-negative bacteria, including carbapenemase-producing strains. Nonetheless, only a few reports are available regarding the acquisition of resistance in clinical settings, primarily due to its recent usage. This study aimed to investigate cefiderocol resistance using an in vitro resistance development model to gain insights into the underlying molecular resistance mechanisms. Cefiderocol susceptible reference strains (Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa) and a clinical Acinetobacter baumannii complex isolate were exposed to increasing cefiderocol concentrations using a high-throughput resistance development model. Cefiderocol susceptibility testing was performed using broth microdilution. Whole-genome sequencing was employed to identify newly acquired resistance mutations. Our in vitro resistance development model led to several clones of strains exhibiting cefiderocol resistance, with MIC values 8-fold to 512-fold higher than initial levels. In total, we found 42 different mutations in 26 genes, of which 35 could be described for the first time. Putative loss-of-function mutations were detected in the envZ, tonB, and cirA genes in 13 out of 17 isolates, leading to a decrease in cefiderocol influx. Other potential resistance mechanisms included multidrug efflux pumps (baeS, czcS, nalC), antibiotic-inactivating enzymes (ampR, dacB), and target mutations in penicillin-binding-protein genes (mrcB). This study reveals new insights into underlying molecular resistance mechanisms against cefiderocol. While mutations leading to reduced influx via iron transporters was the most frequent resistance mechanism, we also detected several other novel resistance mutations causing cefiderocol resistance.
Collapse
Affiliation(s)
- Richard Kriz
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Section Biomedical Science, Health Sciences, FH Campus Wien University of Applied Sciences, Vienna, Austria
| | - Kathrin Spettel
- Section Biomedical Science, Health Sciences, FH Campus Wien University of Applied Sciences, Vienna, Austria
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Alina Pichler
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
- Section Biomedical Science, Health Sciences, FH Campus Wien University of Applied Sciences, Vienna, Austria
- Pediatric Laboratory, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Katharina Schefberger
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Maria Sanz-Codina
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Felix Lötsch
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Nicole Harrison
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Birgit Willinger
- Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Heinz Burgmann
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Heimo Lagler
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
35
|
Jia X, Zhu Y, Jia P, Li C, Chu X, Sun T, Liu X, Yu W, Chen F, Xu Y, Yang Q. The key role of iroBCDN-lacking pLVPK-like plasmid in the evolution of the most prevalent hypervirulent carbapenem-resistant ST11-KL64 Klebsiella pneumoniae in China. Drug Resist Updat 2024; 77:101137. [PMID: 39178714 DOI: 10.1016/j.drup.2024.101137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/31/2024] [Accepted: 08/11/2024] [Indexed: 08/26/2024]
Abstract
AIMS Hypervirulent carbapenem-resistant Klebsiella pneumoniae (hv-CRKP), coharboring hypervirulence and carbapenem-resistance genes mediated by plasmids, causes infections with extremely high mortality and seriously impacts public health. Exploring the transfer mechanisms of virulence/carbapenem-resistance plasmids, as well as the formation and evolution pathway of hv-CRKP is of great significance to the control of hv-CRKP infections. METHODS In this study, we identified the predominant clone of hv-CRKP in China and elucidated its genomic characteristics and formation route based on 239 multicenter clinical K. pneumoniae isolates and 1014 GenBank genomes by using comparative genomic analysis. Further, we revealed the factors affecting the transfer of virulence plasmids, and explained the genetic foundation for the prevalence of Chinese predominant hv-CRKP clone. RESULTS ST11-KL64 is the predominant clone of hv-CRKP in China and primarily evolved from ST11-KL64 CRKP by acquiring the pLVPK-like virulence plasmid from hvKP. Significantly, the virulence gene cluster iroBCDN was lost in the virulence plasmid of ST11-KL64 hv-CRKP but existed in that of hvKP. Moreover, the absence of iroBCDN didn't decrease the virulence of hv-CRKP, which was proved by bacterial test, cell-interaction test and mice infection model. On the contrary, loss of iroBCDN was observed to regulate virulence/carbapenem-resistance plasmid transfer and oxidative stress-related genes in strains and thus promoted the mobilization of nonconjugative virulence plasmid from hvKP into ST11-KL64 CRKP, forming hv-CRKP which finally had elevated antioxidant capacity and enhanced survival capacity in macrophages. The loss of iroBCDN increased the survival ability of hv-CRKP without decreasing its virulence, endowing it with an evolutionary advantage. CONCLUSIONS Our work provides new insights into the key role of iroBCDN loss in convergence of CRKP and hvKP, and the genetic and biological foundation for the widespread prevalence of ST11-KL64 hv-CRKP in China.
Collapse
Affiliation(s)
- Xinmiao Jia
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Center for bioinformatics, National Infrastructures for Translational Medicine, Institute of Clinical Medicine & Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Zhu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Peiyao Jia
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cuidan Li
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Xiaobing Chu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Tianshu Sun
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Clinical Biobank, Center for Biomedical Technology, National Science and Technology Key Infrastructure on Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyu Liu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Yu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Graduate School, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Chen
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yingchun Xu
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiwen Yang
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing, China.
| |
Collapse
|
36
|
Kondaboina S, Parrish O, Parada CA, Ferreira M. Whole Exome Sequencing of Intracranial Epidermoid Cysts Reveals Immune-Associated Mechanistic and Potential Targets. Cancers (Basel) 2024; 16:3487. [PMID: 39456581 PMCID: PMC11506683 DOI: 10.3390/cancers16203487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Intracranial Epidermoid Cysts (IECs) are rare intracranial tumors primarily treated through surgery. Cyst adherence complicates complete removal, leading to high rates of tumor progression after subtotal resection. The molecular drivers of IEC remain unknown. Consequently, advances in treatment have fallen short. Tumor genetic profiling has revealed potential targets for drug development, including FDA-approved options and reshaping treatment. The genetic landscape of IECs has not been explored. We applied Whole Exome Sequencing (WES) to IECs to gain insights into the mechanisms of oncogenesis and identify potential therapeutic targets. Methods: We performed WES on tumor tissue and matched blood samples, when available. Following GATK best practices, we conducted read processing, quality control, somatic variant calling, and copy-number inference. Data analyses and visualization were conducted in R. Results: Top altered genes are associated with the immune system and tumor microenvironment, suggesting a mechanism of immune evasion. Gene and pathway enrichment revealed a high mutation burden in genes associated with Extracellular Matrix (ECM) and PI3K-AKT-mTOR cascades. Recurrent and deleterious alterations in NOTCH2 and USP8 were identified in 50% and 30% of the cohort, respectively. Frequent amplifications in deubiquitinases and beta-defensins strengthened the involvement of immune mechanisms for oncogenic transformation. Conclusions: Top altered genes and recurrent mutations may play a role in shaping the microenvironment and modulating immune evasion in IECs. USP8 and NOTCH2 may serve as clinically relevant target for IECs. Finally, we present evidence that the crosstalk between the PI3K-Akt-mTOR and ECM signaling pathways may play a role in modulating the immune escape mechanism in IECs.
Collapse
Affiliation(s)
| | | | - Carolina Angelica Parada
- Department of Neurological Surgery, University of Washington Medical Center 1, Seattle, WA 98195, USA; (S.K.); (O.P.)
| | - Manuel Ferreira
- Department of Neurological Surgery, University of Washington Medical Center 1, Seattle, WA 98195, USA; (S.K.); (O.P.)
| |
Collapse
|
37
|
Mogrovejo‐Arias DC, Hay MC, Edwards A, Mitchell AC, Steinmann J, Brill FHH, Neumann B. Investigating the resistome of haemolytic bacteria in Arctic soils. ENVIRONMENTAL MICROBIOLOGY REPORTS 2024; 16:e70028. [PMID: 39440916 PMCID: PMC11497493 DOI: 10.1111/1758-2229.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Microorganisms inhabiting hostile Arctic environments express a variety of functional phenotypes, some of clinical interest, such as haemolytic ability and antimicrobial resistance. We studied haemolytic bacterial isolates from Arctic habitats, assessing their minimum inhibitory concentration (MIC) against antimicrobials. We then performed whole genome sequencing and analysed them for features conferring antimicrobial resistance. MIC data showed that Micromonospora spp. belong to 33% non-wild type (NWT) for erythromycin and penicillin and 22% NWT for tetracycline. Both Pseudomonas spp. belong to 43% NWT for nalidixic acid and streptomycin and 29% NWT for colistin. Finally, the Pedobacter isolate was in 80% NWT for antimicrobials tested. Whole-genome sequencing analyses revealed that fluoroquinolones, tetracyclines, macrolides and penams were the most frequent drug classes against which genotypic resistance was found. Additionally, resistance genes to heavy metals and disinfectants were identified. Our research demonstrates the presence of antimicrobial resistance in bacteria from Arctic habitats and highlights the importance of conservation efforts in these environments, where anthropogenic influence is becoming more evident. Furthermore, our data suggest the possible presence of novel resistance mechanisms, which could pose a threat if the responsible genes are transferable between species or become widespread due to environmental stress and alterations brought about by climate change.
Collapse
Affiliation(s)
- Diana C. Mogrovejo‐Arias
- MicroArctic Research, Dr. Brill + Partner GmbH Institut für Hygiene und MikrobiologieHamburgGermany
| | - Melanie C. Hay
- Institute of Biological, Environmental & Rural Sciences (IBERS)Aberystwyth UniversityAberystwythUK
- Department of Pathobiology and Population SciencesThe Royal Veterinary CollegeBrookmans ParkUK
| | - Arwyn Edwards
- Institute of Biological, Environmental & Rural Sciences (IBERS)Aberystwyth UniversityAberystwythUK
| | - Andrew C. Mitchell
- Department of Geography and Earth SciencesAberystwyth UniversityAberystwythUK
| | - Jörg Steinmann
- Institute of Clinical Microbiology, Infectious Diseases and Infection ControlParacelsus Medical University, Klinikum NürnbergNurembergGermany
| | - Florian H. H. Brill
- MicroArctic Research, Dr. Brill + Partner GmbH Institut für Hygiene und MikrobiologieHamburgGermany
| | - Bernd Neumann
- Institute of Clinical Microbiology, Infectious Diseases and Infection ControlParacelsus Medical University, Klinikum NürnbergNurembergGermany
| |
Collapse
|
38
|
Fleskes RE, Johnson SJ, Honap TP, Abin CA, Gilmore JK, Oubré L, Bueschgen WD, Abel SM, Ofunniyin AA, Lewis CM, Schurr TG. Oral microbial diversity in 18th century African individuals from South Carolina. Commun Biol 2024; 7:1213. [PMID: 39342044 PMCID: PMC11439080 DOI: 10.1038/s42003-024-06893-0] [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: 11/15/2023] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
As part of the Anson Street African Burial Ground Project, we characterized the oral microbiomes of twelve 18th century African-descended individuals (Ancestors) from Charleston, South Carolina, USA, to study their oral health and diet. We found that their oral microbiome composition resembled that of other historic (18th-19th century) dental calculus samples but differed from that of modern samples, and was not influenced by indicators of oral health and wear observed in the dentition. Phylogenetic analysis of the oral bacteria, Tannerella forsythia and Pseudoramibacter alactolyticus, revealed varied patterns of lineage diversity and replacement in the Americas, with the Ancestors carrying strains similar to historic period Europeans and Africans. Functional profiling of metabolic pathways suggested that the Ancestors consumed a diet low in animal protein. Overall, our study reveals important insights into the oral microbial histories of African-descended individuals, particularly oral health and diet in colonial North American enslavement contexts.
Collapse
Affiliation(s)
- Raquel E Fleskes
- Department of Anthropology, Dartmouth College, Hanover, NH, USA.
- The Anson Street African Burial Ground Project, Mount Pleasant, SC, USA.
| | - Sarah J Johnson
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, OK, USA
- Department of Anthropology, University of Oklahoma, Norman, OK, USA
| | - Tanvi P Honap
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, OK, USA
- Department of Anthropology, University of Oklahoma, Norman, OK, USA
| | - Christopher A Abin
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, OK, USA
- Department of Anthropology, University of Oklahoma, Norman, OK, USA
| | - Joanna K Gilmore
- The Anson Street African Burial Ground Project, Mount Pleasant, SC, USA
- Department of Sociology and Anthropology, College of Charleston, Charleston, SC, USA
| | - La'Sheia Oubré
- The Anson Street African Burial Ground Project, Mount Pleasant, SC, USA
| | | | - Suzanne M Abel
- Charleston County Coroner's Office, North Charleston, SC, USA
| | - Ade A Ofunniyin
- The Anson Street African Burial Ground Project, Mount Pleasant, SC, USA
- Department of Sociology and Anthropology, College of Charleston, Charleston, SC, USA
| | - Cecil M Lewis
- Laboratories of Molecular Anthropology and Microbiome Research (LMAMR), University of Oklahoma, Norman, OK, USA.
- Department of Anthropology, University of Oklahoma, Norman, OK, USA.
| | - Theodore G Schurr
- The Anson Street African Burial Ground Project, Mount Pleasant, SC, USA.
- Department of Anthropology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
39
|
Bhoyar RC, Jolly B, Vignesh H, Bhatt L, Senthivel V, Israni R, Scaria V, Sivasubbu S. Protocol for next-generation sequencing of the LSD virus genome using an amplicon-based approach. STAR Protoc 2024; 5:103020. [PMID: 39083382 PMCID: PMC11342167 DOI: 10.1016/j.xpro.2024.103020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/03/2023] [Accepted: 03/29/2024] [Indexed: 08/02/2024] Open
Abstract
Lumpy skin disease (LSD) is a viral disease predominantly affecting cattle caused by a poxvirus belonging to the capripoxvirus genus. Here, we present a protocol for next-generation sequencing of the LSD virus genome using an amplicon-based approach. We describe steps for DNA extraction, viral DNA enrichment, amplicon pooling and purification, and library preparation and pooling. We then detail procedures for sequencing and computational analysis. This protocol can be adapted to any Illumina sequencing platform as an accelerated and scalable system. For complete details on the use and execution of this protocol, please refer to Bhatt et al.1,2.
Collapse
Affiliation(s)
- Rahul C Bhoyar
- CSIR- Institute of Genomics and Integrative Biology (CSIR-IGIB), Mathura Road, Delhi 110025, India
| | - Bani Jolly
- CSIR- Institute of Genomics and Integrative Biology (CSIR-IGIB), Mathura Road, Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Harie Vignesh
- CSIR- Institute of Genomics and Integrative Biology (CSIR-IGIB), Mathura Road, Delhi 110025, India
| | - Lenin Bhatt
- State Disease Diagnostic Centre, Department of Animal Husbandry, Jaipur, Rajasthan 302001, India
| | - Vigneshwar Senthivel
- CSIR- Institute of Genomics and Integrative Biology (CSIR-IGIB), Mathura Road, Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Ravi Israni
- State Disease Diagnostic Centre, Department of Animal Husbandry, Jaipur, Rajasthan 302001, India
| | - Vinod Scaria
- CSIR- Institute of Genomics and Integrative Biology (CSIR-IGIB), Mathura Road, Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| | - Sridhar Sivasubbu
- CSIR- Institute of Genomics and Integrative Biology (CSIR-IGIB), Mathura Road, Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| |
Collapse
|
40
|
Wiens M, Farahani H, Scott RW, Underhill TM, Bashashati A. Benchmarking bulk and single-cell variant-calling approaches on Chromium scRNA-seq and scATAC-seq libraries. Genome Res 2024; 34:1196-1210. [PMID: 39147582 PMCID: PMC11444184 DOI: 10.1101/gr.277066.122] [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: 08/05/2023] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Single-cell sequencing methodologies such as scRNA-seq and scATAC-seq have become widespread and effective tools to interrogate tissue composition. Increasingly, variant callers are being applied to these methodologies to resolve the genetic heterogeneity of a sample, especially in the case of detecting the clonal architecture of a tumor. Typically, traditional bulk DNA variant callers are applied to the pooled reads of a single-cell library to detect candidate mutations. Recently, multiple studies have applied such callers on reads from individual cells, with some citing the ability to detect rare variants with higher sensitivity. Many studies apply these two approaches to the Chromium (10x Genomics) scRNA-seq and scATAC-seq methodologies. However, Chromium-based libraries may offer additional challenges to variant calling compared with existing single-cell methodologies, raising questions regarding the validity of variants obtained from such a workflow. To determine the merits and challenges of various variant-calling approaches on Chromium scRNA-seq and scATAC-seq libraries, we use sample libraries with matched bulk whole-genome sequencing to evaluate the performance of callers. We review caller performance, finding that bulk callers applied on pooled reads significantly outperform individual-cell approaches. We also evaluate variants unique to scRNA-seq and scATAC-seq methodologies, finding patterns of noise but also potential capture of RNA-editing events. Finally, we review the notion that variant calling at the single-cell level can detect rare somatic variants, providing empirical results that suggest resolving such variants is infeasible in single-cell Chromium libraries.
Collapse
Affiliation(s)
- Matthew Wiens
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 2B9, Canada
| | - Hossein Farahani
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 2B9, Canada
| | - R Wilder Scott
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 2B9, Canada
| | - T Michael Underhill
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 2B9, Canada
- Department of Cellular & Physiological Sciences, University of British Columbia, Vancouver, British Columbia V6T 2A1, Canada
| | - Ali Bashashati
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 2B9, Canada;
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, British Columbia V6T 1Z7, Canada
| |
Collapse
|
41
|
Xu J, Wan R, Cai Y, Cai S, Wu L, Li B, Duan J, Cheng Y, Li X, Wang X, Han L, Wu X, Fan Y, Yu Y, Lv D, Shi J, Huang J, Zhou S, Han B, Sun G, Guo Q, Ji Y, Zhu X, Hu S, Zhang W, Wang Q, Jia Y, Wang Z, Song Y, Wu J, Shi M, Li X, Han Z, Liu Y, Yu Z, Liu AW, Wang X, Zhou C, Zhong D, Miao L, Zhang Z, Zhao H, Yang J, Wang D, Wang Y, Li Q, Zhang X, Ji M, Yang Z, Cui J, Gao B, Wang B, Liu H, Nie L, He M, Jin S, Gu W, Shu Y, Zhou T, Feng J, Yang X, Huang C, Zhu B, Yao Y, Yu J, Yao S, Shen R, Wang Z, Wang J. Circulating tumor DNA-based stratification strategy for chemotherapy plus PD-1 inhibitor in advanced non-small-cell lung cancer. Cancer Cell 2024; 42:1598-1613.e4. [PMID: 39255777 DOI: 10.1016/j.ccell.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/19/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
Stratification strategies for chemotherapy plus PD-1 inhibitors in advanced non-small-cell lung cancer (NSCLC) are critically demanded. We performed high-throughput panel-based deep next-generation sequencing and low-pass whole genome sequencing on prospectively collected circulating tumor DNA (ctDNA) specimens from 460 patients in the phase 3 CHOICE-01 study at different time points. We identified predictive markers for chemotherapy plus PD-1 inhibitor, including ctDNA status and genomic features such as blood-based tumor mutational burden, intratumor heterogeneity, and chromosomal instability. Furthermore, we established an integrated ctDNA-based stratification strategy, blood-based genomic immune subtypes (bGIS) scheme, to distinguish patients who benefit from the addition of PD-1 inhibitor to first-line chemotherapy. Moreover, we demonstrated potential applications for the dynamic monitoring of ctDNA. Overall, we proposed a potential therapeutic algorithm based on the ctDNA-based stratification strategy, shedding light on the individualized management of immune-chemotherapies for patients with advanced NSCLC.
Collapse
Affiliation(s)
- Jiachen Xu
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Wan
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiran Cai
- Burning Rock Biotech, Guangzhou, China
| | | | - Lin Wu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Baolan Li
- Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jianchun Duan
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | - Xiaoling Li
- Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Xicheng Wang
- The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Liang Han
- Xuzhou Central Hospital, Xuzhou, China
| | - Xiaohong Wu
- Jiangnan University Affiliated Hospital, Wuxi, China
| | - Yun Fan
- Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
| | - Yan Yu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Dongqing Lv
- Taizhou Hospital of Zhejiang Province, Linhai, China
| | | | - Jianjin Huang
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shaozhang Zhou
- Guangxi Medical University Affiliated Tumor Hospital, Nanning, China
| | - Baohui Han
- Shanghai Chest Hospital, Shanghai, China
| | - Guogui Sun
- Tangshan People's Hospital, Tangshan, China
| | - Qisen Guo
- Shangdong Cancer Hospital, Jinan, China
| | - Youxin Ji
- Qingdao Central Hospital, Qingdao, China
| | - Xiaoli Zhu
- Zhongda Hospital Southeast University, Nanjing, China
| | - Sheng Hu
- Hubei Cancer Hospital, Wuhan, China
| | - Wei Zhang
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | | | - Yuming Jia
- The Second People's Hospital of Yibin, Yibin, China
| | - Ziping Wang
- Peking University Cancer Hospital, Beijing, China
| | - Yong Song
- Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jingxun Wu
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Meiqi Shi
- Jiangsu Cancer Hospital, Nanjing, China
| | - Xingya Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Han
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Yunpeng Liu
- The First Hospital of China Medical University, Shenyang, China
| | - Zhuang Yu
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - An-Wen Liu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiuwen Wang
- Qilu Hospital of Shandong University, Jinan, China
| | - Caicun Zhou
- Shanghai Pulmonary Hospital, Shanghai, China
| | | | - Liyun Miao
- Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | | | - Hui Zhao
- The Second Hospital of Anhui Medical University, Hefei, China
| | - Jun Yang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dong Wang
- Army Medical Center of PLA, Daping Hospital, Daping, China
| | - Yingyi Wang
- Peking Union Medical College Hospital, Beijing, China
| | - Qiang Li
- Shanghai East Hospital of Tongji University, Shanghai, China
| | | | - Mei Ji
- The First People's Hospital of Changzhou, Changzhou, China
| | - Zhenzhou Yang
- The Second Affiliated Hospital of Chongqing University, Chongqing, China
| | - Jiuwei Cui
- The First Hospital of Jilin University, Jilin, China
| | - Beili Gao
- Ruijin Hospital Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Buhai Wang
- Subei People's Hospital of Jiangsu Province, Yanghzou, China
| | - Hu Liu
- Anhui Provincial Cancer Hospital, Hefei, China
| | - Lei Nie
- Shanxi Provincial Tumor Hospital, Xian, China
| | - Mei He
- Shanxi Provincial People's Hospital, Taiyuan, China
| | - Shi Jin
- Cancer Hospital of Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, China
| | - Wei Gu
- Nanjing First Hospital, Nanjing, China
| | - Yongqian Shu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tong Zhou
- ChangZhou Cancer Hospital, Changzhou, China
| | - Jian Feng
- Affiliated Hospital of Nantong University, Nantong, China
| | | | | | - Bo Zhu
- Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Yu Yao
- First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Jianjun Yu
- Shanghai Junshi Biosciences, Shanghai, China
| | - Sheng Yao
- Shanghai Junshi Biosciences, Shanghai, China
| | | | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
42
|
Huang J, Mao L, Lei Q, Guo AY. Bioinformatics tools and resources for cancer and application. Chin Med J (Engl) 2024; 137:2052-2064. [PMID: 39075637 PMCID: PMC11374212 DOI: 10.1097/cm9.0000000000003254] [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: 04/20/2024] [Indexed: 07/31/2024] Open
Abstract
ABSTRACT Tumor bioinformatics plays an important role in cancer research and precision medicine. The primary focus of traditional cancer research has been molecular and clinical studies of a number of fundamental pathways and genes. In recent years, driven by breakthroughs in high-throughput technologies, large-scale cancer omics data have accumulated rapidly. How to effectively utilize and share these data is particularly important. To address this crucial task, many computational tools and databases have been developed over the past few years. To help researchers quickly learn and understand the functions of these tools, in this review, we summarize publicly available bioinformatics tools and resources for pan-cancer multi-omics analysis, regulatory analysis of tumorigenesis, tumor treatment and prognosis, immune infiltration analysis, immune repertoire analysis, cancer driver gene and driver mutation analysis, and cancer single-cell analysis, which may further help researchers find more suitable tools for their research.
Collapse
Affiliation(s)
- Jin Huang
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lingzi Mao
- Hubei Bioinformatics & Molecular Imaging Key Laboratory, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei 430074, China
| | - Qian Lei
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - An-Yuan Guo
- Department of Thoracic Surgery, West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
43
|
Mariner-Llicer C, Goig GA, Torres-Puente M, Vashakidze S, Villamayor LM, Saavedra-Cervera B, Mambuque E, Khurtsilava I, Avaliani Z, Rosenthal A, Gabrielian A, Shurgaia M, Shubladze N, García-Basteiro AL, López MG, Comas I. Genetic diversity within diagnostic sputum samples is mirrored in the culture of Mycobacterium tuberculosis across different settings. Nat Commun 2024; 15:7114. [PMID: 39237504 PMCID: PMC11377819 DOI: 10.1038/s41467-024-51266-0] [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: 03/06/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024] Open
Abstract
Culturing and genomic sequencing of Mycobacterium tuberculosis (MTB) from tuberculosis (TB) cases is the basis for many research and clinical applications. The alternative, culture-free sequencing from diagnostic samples, is promising but poses challenges to obtain and analyse the MTB genome. Paradoxically, culture is assumed to impose a diversity bottleneck, which, if true, would entail unexplored consequences. To unravel this paradox we generate high-quality genomes of sputum-culture pairs from two different settings after developing a workflow for sequencing from sputum and a tailored bioinformatics analysis. Careful downstream comparisons reveal sources of sputum-culture incongruences due to false positive/negative variation associated with factors like low input MTB DNA or variable genomic depths. After accounting for these factors, contrary to the bottleneck dogma, we identify a 97% variant agreement within sputum-culture pairs, with a high correlation also in the variants' frequency (0.98). The combined analysis from five different settings and more than 100 available samples shows that our results can be extrapolated to different TB epidemic scenarios, demonstrating that for the cases tested culture accurately mirrors clinical samples.
Collapse
Affiliation(s)
| | - Galo A Goig
- University of Basel, Basel, Switzerland
- Swiss Tropical and Public Health Institute, Allschwil, Switzerland
| | | | - Sergo Vashakidze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
- The University of Georgia, Tbilisi, Georgia
| | - Luis M Villamayor
- FISABIO, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana, València, Spain
| | - Belén Saavedra-Cervera
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- Wellcome Sanger Institute, Hinxton, UK
| | - Edson Mambuque
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Iza Khurtsilava
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Zaza Avaliani
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
- European University, Tbilisi, Georgia
| | - Alex Rosenthal
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Andrei Gabrielian
- Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Marika Shurgaia
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Natalia Shubladze
- National Center for Tuberculosis and Lung Diseases, Tbilisi, Georgia
| | - Alberto L García-Basteiro
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- CIBERINFEC, Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Barcelona, Spain
| | - Mariana G López
- Instituto de Biomedicina de Valencia, IBV, CSIC, València, Spain.
| | - Iñaki Comas
- Instituto de Biomedicina de Valencia, IBV, CSIC, València, Spain.
- CIBERESP, Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública, Madrid, Spain.
| |
Collapse
|
44
|
Fernandez MM, Yu L, Jia Q, Wang X, Hart KL, Jia Z, Lin RJ, Wang L. Engineering Oncogenic Hotspot Mutations on SF3B1 via CRISPR-Directed PRECIS Mutagenesis. CANCER RESEARCH COMMUNICATIONS 2024; 4:2498-2513. [PMID: 39194178 PMCID: PMC11421219 DOI: 10.1158/2767-9764.crc-24-0145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/01/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
SF3B1 is the most recurrently mutated RNA splicing gene in cancer. However, research of its pathogenic role has been hindered by a lack of disease-relevant cell line models. Here, our study compared four genome engineering platforms to establish SF3B1 mutant cell lines: CRISPR-Cas9 editing, AAV homology-directed repair editing, base editing (ABEmax, ABE8e), and prime editing (PE2, PE3, PE5max). We showed that prime editing via PE5max achieved the most efficient SF3B1 K700E editing across a wide range of cell lines. Our approach was further refined by coupling prime editing with a fluorescent reporter that leverages a SF3B1 mutation-responsive synthetic intron to mark successfully edited cells. By applying this approach, called prime editing coupled intron-assisted selection (PRECIS), we introduced the K700E hotspot mutation into two chronic lymphocytic leukemia cell lines, HG-3 and MEC-1. We demonstrated that our PRECIS-engineered cells faithfully recapitulate known mutant SF3B1 phenotypes, including altered splicing, copy number variations, and cell-growth defect. Moreover, we discovered that the SF3B1 mutation can cause the loss of Y chromosome in chronic lymphocytic leukemia. Our results showcase that PRECIS is an efficient and generalizable method for engineering genetically faithful SF3B1 mutant models. Our approach provides new insights on the role of SF3B1 mutation in cancer and enables the generation of SF3B1 mutant cell lines in relevant cellular context. SIGNIFICANCE This study developed an approach that can reliably and efficiently engineer SF3B1 mutation into different cellular contexts, thereby revealing novel roles of SF3B1 mutation in driving aberrant splicing, clonal evolution, and genome instability.
Collapse
Affiliation(s)
- Mike M. Fernandez
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, California.
- Irell & Manella Graduate School of Biological Sciences, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, California.
| | - Lei Yu
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California.
| | - Qiong Jia
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California.
| | - Xuesong Wang
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California.
| | - Kevyn L. Hart
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, California.
| | - Zhenyu Jia
- Department of Botany and Plant Sciences, University of California, Riverside, Riverside, California.
| | - Ren-Jang Lin
- Center for RNA Biology and Therapeutics, Beckman Research Institute, City of Hope, Duarte, California.
| | - Lili Wang
- Department of Systems Biology, Beckman Research Institute, City of Hope National Comprehensive Cancer Center, Monrovia, California.
- Toni Stephenson Lymphoma Center, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Duarte, California.
| |
Collapse
|
45
|
Lombardo M, Camellin U, Gioia R, Serrao S, Scorcia V, Roszkowska AM, Lombardo G, Bertelli M, Medori MC, Alunni Fegatelli D, Vestri A, Mencucci R, Schiano Lomoriello D. Targeted next-generation sequencing analysis in Italian patients with keratoconus. Eye (Lond) 2024; 38:2610-2618. [PMID: 38684849 PMCID: PMC11383948 DOI: 10.1038/s41433-024-03090-5] [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: 06/18/2023] [Revised: 03/12/2024] [Accepted: 04/12/2024] [Indexed: 05/02/2024] Open
Abstract
OBJECTIVE To report variants in 26 candidate genes and describe the clinical features of Italian patients with keratoconus (KC). SUBJECTS/METHODS Sixty-four patients with a confirmed diagnosis of KC were enrolled in this genetic association study. Patients were classified into two study groups according to whether they had a confirmed diagnosis of progressive or stable KC. A purpose-developed Next Generation Sequencing (NGS) panel was used to identify and analyse the coding exons and flanking exon/intron boundaries of 26 genes known to be associated with KC and corneal dystrophies. Interpretation of the pathogenic significance of variants was performed using in silico predictive algorithms. RESULT The targeted NGS research identified a total of 167 allelic variants of 22 genes in the study population; twenty-four patients had stable keratoconus (n. 54 variants) and forty patients had progressive disease (n. 113 variants). We identified genetic variants of certain pathogenic significance in five patients with progressive KC; in addition, eight novel genetic variants were found in eight patients with progressive KC. Mutations of FLG, LOXHD1, ZNF469, and DOCK9 genes were twice more frequently identified in patients with progressive than stable disease. Filaggrin gene variants were found in 49 patients (76% of total), of whom 32 patients (80% of progressive KC group) had progressive disease. CONCLUSIONS Targeted NGS research provided new insights into the causative effect of candidate genes in the clinical phenotype of keratoconus. Filaggrin mutations were found to represent a genetic risk factor for development of progressive disease in Italy.
Collapse
Affiliation(s)
- Marco Lombardo
- Studio Italiano di Oftalmologia, Via Livenza 3, 00198, Rome, Italy.
- Vision Engineering Italy srl, Via Livenza 3, 00198, Rome, Italy.
| | - Umberto Camellin
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Via Consolare Valeria 1, 98124, Messina, Italy
| | - Raffaella Gioia
- Department of Ophthalmology, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Sebastiano Serrao
- Studio Italiano di Oftalmologia, Via Livenza 3, 00198, Rome, Italy
- Vision Engineering Italy srl, Via Livenza 3, 00198, Rome, Italy
| | - Vincenzo Scorcia
- Department of Ophthalmology, University Magna Graecia of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Anna Maria Roszkowska
- Department of Biomedical Sciences, Ophthalmology Clinic, University of Messina, Via Consolare Valeria 1, 98124, Messina, Italy
- Ophthalmology Department, Faculty of Medicine and Health Sciences, Andrzej Frycz Modrzewski University, Krakow, Poland
| | - Giuseppe Lombardo
- Vision Engineering Italy srl, Via Livenza 3, 00198, Rome, Italy
- CNR-IPCF, Istituto per i Processi Chimico-Fisici, Viale F. Stagno D'Alcontres 37, 98158, Messina, Italy
| | | | - Maria Chiara Medori
- MAGI's Lab srl, Via Maioliche 57, 38068, Rovereto, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, via A. Moro 2, Siena, Italy
| | - Danilo Alunni Fegatelli
- Department of Public Health and infectious diseases, University of Rome "La Sapienza", Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Annarita Vestri
- Department of Public Health and infectious diseases, University of Rome "La Sapienza", Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Rita Mencucci
- Ophthalmology Clinic, AOU Careggi, University of Florence, Largo Brambilla 3, 50134, Firenze, Italy
| | | |
Collapse
|
46
|
Premanand A, Shanmuga Priya M, Reena Rajkumari B. Genetic variants in androgenetic alopecia: insights from scalp RNA sequencing data. Arch Dermatol Res 2024; 316:590. [PMID: 39215850 DOI: 10.1007/s00403-024-03351-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 08/03/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Affiliation(s)
- A Premanand
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - M Shanmuga Priya
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - B Reena Rajkumari
- Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
47
|
Giovannini S, Smirnov A, Concetti L, Scimeca M, Mauriello A, Bischof J, Rovella V, Melino G, Buonomo CO, Candi E, Bernassola F. A comprehensive molecular characterization of a claudin-low luminal B breast tumor. Biol Direct 2024; 19:66. [PMID: 39152485 PMCID: PMC11328405 DOI: 10.1186/s13062-024-00482-1] [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: 04/23/2024] [Accepted: 05/20/2024] [Indexed: 08/19/2024] Open
Abstract
Breast cancer is the most common cause of death from cancer in women. Here, we present the case of a 43-year-old woman, who received a diagnosis of claudin-low luminal B breast cancer. The lesion revealed to be a poorly differentiated high-grade infiltrating ductal carcinoma, which was strongly estrogen receptor (ER)/progesterone receptor (PR) positive and human epidermal growth factor receptor (HER2) negative. Her tumor underwent in-depth chromosomal, mutational and gene expression analyses. We found a pathogenic protein truncating mutation in the TP53 gene, which is predicted to disrupt its transcriptional activity. The patient also harbors germline mutations in some mismatch repair (MMR) genes, and her tumor displays the presence of immune infiltrates, high tumor mutational burden (TMB) status and the apolipoprotein B mRNA editing enzyme catalytic polypeptide 3 (APOBEC3) associated signatures, which, overall, are predictive for the use of immunotherapy. Here, we propose promising prognostic indicators as well as potential therapeutic strategies based on the molecular characterization of the tumor.
Collapse
Affiliation(s)
- Sara Giovannini
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Artem Smirnov
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
- Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy
| | - Livia Concetti
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Manuel Scimeca
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Julia Bischof
- Germany Biochemistry Laboratory, Indivumed GmbH, Falkenried, 88 Building D, 20251, Hamburg, Germany
| | - Valentina Rovella
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Claudio Oreste Buonomo
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| | - Eleonora Candi
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
- Istituto Dermopatico Immacolata (IDI-IRCCS), 00100, Rome, Italy.
| | - Francesca Bernassola
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, 00133, Rome, Italy.
| |
Collapse
|
48
|
Rakshit P, Singh A, Singh R, Banerjee T. An in-depth study on survival mechanism of bacterial isolates in disinfectants within the hospital environment. Front Cell Infect Microbiol 2024; 14:1442914. [PMID: 39211793 PMCID: PMC11358120 DOI: 10.3389/fcimb.2024.1442914] [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: 06/03/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction The emergence of disinfectant resistance has become a severe threat due to reduced effectiveness. This study was undertaken to determine how bacteria adapt to survive exposure to disinfectants in the busiest section of a tertiary care hospital in Varanasi, India. Methods Four isolates (two Klebsiella pneumoniae, Kp1 and Kp2; two Pseudomonas aeruginosa, Pa1 and Pa2) were obtained from chlorhexidine (CHX)-based handwash during microbiological surveillance of "in-use disinfectants" in hospital. Six disinfectants [4% CHX, 2% glutaraldehyde, 7.5% hydrogen peroxide, 1% sodium hypochlorite and 0.1% benzalkonium chloride (BAC), and 70% ethyl alcohol] were tested against these four isolates to determine minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC). Antibiotic profile, change in MIC on exposure to disinfectants and biofilm formation in the presence and absence of disinfectants was studied. Whole genome sequencing (WGS) was done to identify the resistance mechanisms. Result The isolates showed the highest MBC/MIC ratio (4) against glutaraldehyde. Exposure to supra-inhibitory concentration of BAC for 21 days resulted in doubling of MIC/MBC. The majority (75%) of the isolates were multidrug resistant. All the isolates were strong biofilm producers. The reduction rate of biofilm formation decreased with an increase in the concentration of disinfectants (p = 0.05 for BAC). WGS revealed multiple AMR genes including bla DIM-1, disinfectant-resistant gene and efflux pump genes. Conclusion The study emphasized the various adaptation strategies of these isolates for survival in disinfectant environment, thus posing a huge challenge for their control in the hospital environment.
Collapse
Affiliation(s)
- Pue Rakshit
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Aradhana Singh
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ravindra Singh
- Trauma Center, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Tuhina Banerjee
- Department of Microbiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| |
Collapse
|
49
|
Zeng Y, Wei R, Bao L, Xue T, Qin Y, Ren M, Bai Q, Yao Q, Yu C, Chen C, Wei P, Yu B, Cao J, Li X, Zhang Q, Zhou X. Characteristics and Clinical Value of MYC , BCL2, and BCL6 Rearrangement Detected by Next-generation Sequencing in DLBCL. Am J Surg Pathol 2024; 48:919-929. [PMID: 38937822 PMCID: PMC11251499 DOI: 10.1097/pas.0000000000002258] [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: 06/29/2024]
Abstract
MYC , BCL2, and BCL6 rearrangements are clinically important events of diffuse large B-cell lymphoma (DLBCL). The ability and clinical value of targeted next-generation sequencing (NGS) in the detection of these rearrangements in DLBCL have not been fully determined. We performed targeted NGS (481-gene-panel) and break-apart FISH of MYC , BCL2, and BCL6 gene regions in 233 DLBCL cases. We identified 88 rearrangements (16 MYC ; 20 BCL2 ; 52 BCL6 ) using NGS and 96 rearrangements (28 MYC ; 20 BCL2 ; 65 BCL6 ) using FISH. The consistency rates between FISH and targeted NGS for the detection of MYC , BCL2, and BCL6 rearrangements were 93%, 97%, and 89%, respectively. FISH-cryptic rearrangements (NGS+/FISH-) were detected in 7 cases (1 MYC ; 3 BCL2 ; 2 BCL6 ; 1 MYC::BCL6 ), mainly caused by small chromosomal insertions and inversions. NGS-/FISH+ were detected in 38 cases (14 MYC ; 4 BCL2 ; 20 BCL6 ).To clarify the cause of the inconsistencies, we selected 17 from the NGS-/FISH+ rearrangements for further whole genome sequencing (WGS), and all 17 rearrangements were detected with break points by WGS. These break points were all located outside the region covered by the probe of targeted NGS, and most (16/17) were located in the intergenic region. These results indicated that targeted NGS is a powerful clinical diagnostics tool for comprehensive MYC , BCL2, and BCL6 rearrangement detection. Compared to FISH, it has advantages in describing the break point distribution, identifying uncharacterized partners, and detecting FISH-cryptic rearrangements. However, the lack of high-sensitivity caused by insufficient probe coverage is the main limitation of the current technology.
Collapse
Affiliation(s)
- Yupeng Zeng
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Ran Wei
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Longlong Bao
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Tian Xue
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Yulan Qin
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu
| | - Min Ren
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Qianming Bai
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Qianlan Yao
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Chengli Yu
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Chen Chen
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Baohua Yu
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Junning Cao
- Department of Oncology, Shanghai Medical College
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaoqiu Li
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| | - Qunling Zhang
- Department of Oncology, Shanghai Medical College
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaoyan Zhou
- Department of Pathology, Fudan University Shanghai Cancer Center
- Department of Oncology, Shanghai Medical College
- Institute of Pathology, Fudan University
| |
Collapse
|
50
|
Nagelberg AL, Sihota TS, Chuang YC, Shi R, Chow JLM, English J, MacAulay C, Lam S, Lam WL, Lockwood WW. Integrative genomics identifies SHPRH as a tumor suppressor gene in lung adenocarcinoma that regulates DNA damage response. Br J Cancer 2024; 131:534-550. [PMID: 38890444 PMCID: PMC11300780 DOI: 10.1038/s41416-024-02755-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: 09/20/2023] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Identification of driver mutations and development of targeted therapies has considerably improved outcomes for lung cancer patients. However, significant limitations remain with the lack of identified drivers in a large subset of patients. Here, we aimed to assess the genomic landscape of lung adenocarcinomas (LUADs) from individuals without a history of tobacco use to reveal new genetic drivers of lung cancer. METHODS Integrative genomic analyses combining whole-exome sequencing, copy number, and mutational information for 83 LUAD tumors was performed and validated using external datasets to identify genetic variants with a predicted functional consequence and assess association with clinical outcomes. LUAD cell lines with alteration of identified candidates were used to functionally characterize tumor suppressive potential using a conditional expression system both in vitro and in vivo. RESULTS We identified 21 genes with evidence of positive selection, including 12 novel candidates that have yet to be characterized in LUAD. In particular, SNF2 Histone Linker PHD RING Helicase (SHPRH) was identified due to its frequency of biallelic disruption and location within the familial susceptibility locus on chromosome arm 6q. We found that low SHPRH mRNA expression is associated with poor survival outcomes in LUAD patients. Furthermore, we showed that re-expression of SHPRH in LUAD cell lines with inactivating alterations for SHPRH reduces their in vitro colony formation and tumor burden in vivo. Finally, we explored the biological pathways associated SHPRH inactivation and found an association with the tolerance of LUAD cells to DNA damage. CONCLUSIONS These data suggest that SHPRH is a tumor suppressor gene in LUAD, whereby its expression is associated with more favorable patient outcomes, reduced tumor and mutational burden, and may serve as a predictor of response to DNA damage. Thus, further exploration into the role of SHPRH in LUAD development may make it a valuable biomarker for predicting LUAD risk and prognosis.
Collapse
Affiliation(s)
- Amy L Nagelberg
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tianna S Sihota
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yu-Chi Chuang
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Rocky Shi
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Justine L M Chow
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - John English
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Calum MacAulay
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Stephen Lam
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Wan L Lam
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - William W Lockwood
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada.
- Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|