1
|
Pons C. Qarles: a web server for the quick characterization of large sets of genes. NAR Genom Bioinform 2025; 7:lqaf030. [PMID: 40160219 PMCID: PMC11954521 DOI: 10.1093/nargab/lqaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
The characterization of gene sets is a recurring task in computational biology. Identifying specific properties of a hit set compared to a reference set can reveal biological roles and mechanisms, and can lead to the prediction of new hits. However, collecting the features to evaluate can be time consuming, and implementing an informative but compact graphical representation of the multiple comparisons can be challenging, particularly for bench scientists. Here, I present Qarles (quick characterization of large sets of genes), a web server that annotates Saccharomyces cerevisiae gene sets by querying a database of 31 features widely used by the yeast community and that identifies their specific properties, providing publication-ready figures and reliable statistics. Qarles has a deliberately simple user interface with all the functionality in a single web page and a fast response time to facilitate adoption by the scientific community. Qarles provides a rich and compact graphical output, including up to five gene set comparisons across 31 features in a single dotplot, and interactive boxplots to enable the identification of outliers. Qarles can also predict new hit genes by using a random forest trained on the selected features. The web server is freely available at https://qarles.org.
Collapse
Affiliation(s)
- Carles Pons
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute for Science and Technology (BIST), 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
2
|
Cancer vulnerabilities exposed by finding interactions among DNA repair factors. Nature 2025:10.1038/d41586-025-01049-4. [PMID: 40205104 DOI: 10.1038/d41586-025-01049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
|
3
|
Nadal-Ribelles M, Solé C, Díez-Villanueva A, Stephan-Otto Attolini C, Matas Y, Steinmetz L, de Nadal E, Posas F. A single-cell resolved genotype-phenotype map using genome-wide genetic and environmental perturbations. Nat Commun 2025; 16:2645. [PMID: 40102404 PMCID: PMC11920212 DOI: 10.1038/s41467-025-57600-4] [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: 06/29/2024] [Accepted: 02/14/2025] [Indexed: 03/20/2025] Open
Abstract
Heterogeneity is inherent to living organisms and it determines cell fate and phenotypic variability. Despite its ubiquity, the underlying molecular mechanisms and the genetic basis linking genotype to-phenotype heterogeneity remain a central challenge. Here we construct a yeast knockout library with a clone and genotype RNA barcoding structure suitable for genome-scale analyses to generate a high-resolution single-cell yeast transcriptome atlas of 3500 mutants under control and stress conditions. We find that transcriptional heterogeneity reflects the coordinated expression of specific gene programs, generating a continuous of cell states that can be responsive to external insults. Cell state plasticity can be genetically modulated with mutants that act as state attractors and disruption of state homeostasis results in decreased adaptive fitness. Leveraging on intra-genetic variability, we establish that regulators of transcriptional heterogeneity are functionally diverse and influenced by the environment. Our multimodal perturbation-based single-cell Genotype-to-Transcriptome Atlas in yeast provides insights into organism-level responses.
Collapse
Affiliation(s)
- Mariona Nadal-Ribelles
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Carme Solé
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Anna Díez-Villanueva
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Camille Stephan-Otto Attolini
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Yaima Matas
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Lars Steinmetz
- Department of Genetics, Stanford University, School of Medicine, California, USA
- European Molecular Biology Laboratory, Heidelberg, Germany
| | - Eulàlia de Nadal
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Francesc Posas
- Department of Medicine and Life Sciences, Universitat Pompeu Fabra, Barcelona, Spain.
- Institute for Research in Biomedicine (IRB Barcelona), the Barcelona Institute of Science and Technology, Barcelona, Spain.
| |
Collapse
|
4
|
Smith LA, Cahill JA, Lee JH, Graim K. Equitable machine learning counteracts ancestral bias in precision medicine. Nat Commun 2025; 16:2144. [PMID: 40064867 PMCID: PMC11894161 DOI: 10.1038/s41467-025-57216-8] [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: 02/12/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Gold standard genomic datasets severely under-represent non-European populations, leading to inequities and a limited understanding of human disease. Therapeutics and outcomes remain hidden because we lack insights that could be gained from analyzing ancestrally diverse genomic data. To address this significant gap, we present PhyloFrame, a machine learning method for equitable genomic precision medicine. PhyloFrame corrects for ancestral bias by integrating functional interaction networks and population genomics data with transcriptomic training data. Application of PhyloFrame to breast, thyroid, and uterine cancers shows marked improvements in predictive power across all ancestries, less model overfitting, and a higher likelihood of identifying known cancer-related genes. Validation in fourteen ancestrally diverse datasets demonstrates that PhyloFrame is better able to adjust for ancestry bias across all populations. The ability to provide accurate predictions for underrepresented groups, in particular, is substantially increased. Analysis of performance in the most diverse continental ancestry group, African, illustrates how phylogenetic distance from training data negatively impacts model performance, as well as PhyloFrame's capacity to mitigate these effects. These results demonstrate how equitable artificial intelligence (AI) approaches can mitigate ancestral bias in training data and contribute to equitable representation in medical research.
Collapse
Affiliation(s)
- Leslie A Smith
- Department of Computer & Information Science & Engineering, University of Florida, 1889 Museum Rd, Gainesville, 32611, FL, USA
| | - James A Cahill
- Environmental Engineering Sciences Department, University of Florida, 365 Weil Hall, Gainesville, 32611, FL, USA
- UF Genetics Institute, University of Florida, 2033 Mowry Rd, Gainesville, 32610, FL, USA
| | - Ji-Hyun Lee
- Department of Biostatistics, University of Florida, 2004 Mowry Rd, Gainesville, Gainesville, 32603, FL, USA
- UF Health Cancer Center, University of Florida, 2033 Mowry Rd, Gainesville, 32610, FL, USA
| | - Kiley Graim
- Department of Computer & Information Science & Engineering, University of Florida, 1889 Museum Rd, Gainesville, 32611, FL, USA.
- UF Genetics Institute, University of Florida, 2033 Mowry Rd, Gainesville, 32610, FL, USA.
- UF Health Cancer Center, University of Florida, 2033 Mowry Rd, Gainesville, 32610, FL, USA.
| |
Collapse
|
5
|
Moser SC, Jonkers J. Thirty Years of BRCA1: Mechanistic Insights and Their Impact on Mutation Carriers. Cancer Discov 2025; 15:461-480. [PMID: 40025950 PMCID: PMC11893084 DOI: 10.1158/2159-8290.cd-24-1326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/04/2024] [Accepted: 12/06/2024] [Indexed: 03/04/2025]
Abstract
SIGNIFICANCE Here, we explore the impact of three decades of BRCA1 research on the lives of mutation carriers and propose strategies to improve the prevention and treatment of BRCA1-associated cancer.
Collapse
Affiliation(s)
- Sarah C. Moser
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| | - Jos Jonkers
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Ott DP, Desai S, Solinger JA, Kaech A, Spang A. Coordination between ESCRT function and Rab conversion during endosome maturation. EMBO J 2025; 44:1574-1607. [PMID: 39910226 PMCID: PMC11914609 DOI: 10.1038/s44318-025-00367-7] [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: 04/30/2024] [Revised: 12/12/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
The endosomal pathway is essential for regulating cell signaling and cellular homeostasis. Rab5 positive early endosomes receive proteins from the plasma membrane. Dependent on a ubiquitin mark on the protein, they will be either recycled or sorted into intraluminal vesicles (ILVs) by endosomal sorting complex required for transport (ESCRT) proteins. During endosome maturation Rab5 is replaced by Rab7 on endosomes that are able to fuse with lysosomes to form endolysosomes. However, whether ESCRT-driven ILV formation and Rab5-to-Rab7 conversion are coordinated remains unknown. Here we show that loss of early ESCRTs led to enlarged Rab5 positive endosomes and prohibited Rab conversion. Reduction of ubiquitinated cargo alleviated this phenotype. Moreover, ubiquitinated proteins on the endosomal limiting membrane prevented the displacement of the Rab5 guanine nucleotide exchange factor (GEF) RABX-5 by the GEF for Rab7, SAND-1/CCZ-1. Overexpression of Rab7 could partially overcome this block, even in the absence of SAND-1 or CCZ1, suggesting the presence of a second Rab7 GEF. Our data reveal a hierarchy of events in which cargo corralling by ESCRTs is upstream of Rab conversion, suggesting that ESCRT-0 and ubiquitinated cargo could act as timers that determine the onset of Rab conversion.
Collapse
Affiliation(s)
- Daniel P Ott
- Biozentrum, University of Basel, Basel, Switzerland
| | - Samit Desai
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Andres Kaech
- Center for Microscopy and Image Analysis, University of Zurich, Zürich, Switzerland
| | - Anne Spang
- Biozentrum, University of Basel, Basel, Switzerland.
| |
Collapse
|
7
|
Tran JS, Ward RD, Iruegas-López R, Ebersberger I, Peters JM. Chemical genomics informs antibiotic and essential gene function in Acinetobacter baumannii. PLoS Genet 2025; 21:e1011642. [PMID: 40153700 PMCID: PMC11975115 DOI: 10.1371/journal.pgen.1011642] [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: 12/31/2024] [Revised: 04/07/2025] [Accepted: 03/03/2025] [Indexed: 03/30/2025] Open
Abstract
The Gram-negative pathogen, Acinetobacter baumannii, poses a serious threat to human health due to its role in nosocomial infections that are resistant to treatment with current antibiotics. Despite this, our understanding of fundamental A. baumannii biology remains limited, as many essential genes have not been experimentally characterized. These essential genes are critical for bacterial survival and, thus, represent promising targets for drug discovery. Here, we systematically probe the function of essential genes by screening a CRISPR interference knockdown library against a diverse panel of chemical inhibitors, including antibiotics. We find that most essential genes show chemical-gene interactions, allowing insights into both inhibitor and gene function. For instance, knockdown of lipooligosaccharide (LOS) transport genes increased sensitivity to a broad range of chemicals. Cells with defective LOS transport showed cell envelope hyper-permeability that was dependent on continued LOS synthesis. Using phenotypes across our chemical-gene interaction dataset, we constructed an essential gene network linking poorly understood genes to well-characterized genes in cell division and other processes. Finally, our phenotype-structure analysis identified structurally related antibiotics with distinct cellular impacts and suggested potential targets for underexplored inhibitors. This study advances our understanding of essential gene and inhibitor function, providing a valuable resource for mechanistic studies, therapeutic strategies, and future key targets for antibiotic development.
Collapse
Affiliation(s)
- Jennifer Suzanne Tran
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Ryan David Ward
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Rubén Iruegas-López
- Institute of Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
| | - Ingo Ebersberger
- Institute of Cell Biology and Neuroscience, Goethe University, Frankfurt am Main, Germany
- Senckenberg Biodiversity and Climate Research Centre (S-BIKF), Frankfurt am Main, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Frankfurt am Main, Germany
| | - Jason Matthew Peters
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
8
|
Akoto E, Doss EM, Claypool KP, Owutey SL, Richards KA, Lehman KM, Daraghmi MM, Turk SM, Indovina CJ, Avaala JA, Evans MD, Scott AR, Schneider HO, Rogers EM, True JD, Smaldino PJ, Rubenstein EM. The kinesin Kar3 is required for endoplasmic reticulum-associated degradation. Mol Biol Cell 2025; 36:br9. [PMID: 39841550 PMCID: PMC11974954 DOI: 10.1091/mbc.e24-10-0437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025] Open
Abstract
Degradation of aberrant, excess, and regulatory proteins at the endoplasmic reticulum (ER) is a conserved feature of eukaryotic cells, disruption of which contributes to disease. While remarkable progress has been made in recent years, mechanisms and genetic requirements for ER-associated degradation (ERAD) remain incompletely understood. We recently conducted a screen for genes required for turnover of a model ER translocon-associated substrate of the Hrd1 ubiquitin ligase in Saccharomyces cerevisiae. This screen revealed loss of Kar3 impedes degradation of Deg1*-Sec62, which persistently and aberrantly engages the translocon. Kar3 is a microtubule-associated kinesin 14 family member that impacts multiple aspects of microtubule dynamics during cell division and karyogamy. We investigated involvement of Kar3 and its cofactors in ERAD. Loss of Kar3 hindered ERAD mediated by three ubiquitin ligases but did not impair turnover of a soluble nuclear protein. Further, KAR3 deletion caused hypersensitivity to conditions associated with proteotoxic stress. Kar3's cytoplasmic cofactor Vik1 was also required for efficient degradation of Deg1*-Sec62. Our results reveal a profound and underappreciated role for microtubule-associated proteins in ERAD.
Collapse
Affiliation(s)
- Emmanuel Akoto
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Ellen M. Doss
- Department of Biology, Ball State University, Muncie, IN 47306
| | | | | | | | - Katie M. Lehman
- Department of Biology, Ball State University, Muncie, IN 47306
| | | | | | | | - James A. Avaala
- Department of Biology, Ball State University, Muncie, IN 47306
| | | | | | | | - Evan M. Rogers
- Department of Biology, Ball State University, Muncie, IN 47306
| | - Jason D. True
- Department of Biology, Ball State University, Muncie, IN 47306
| | | | | |
Collapse
|
9
|
Cantore T, Gasperini P, Bevilacqua R, Ciani Y, Sinha S, Ruppin E, Demichelis F. PRODE recovers essential and context-essential genes through neighborhood-informed scores. Genome Biol 2025; 26:42. [PMID: 40022167 PMCID: PMC11869679 DOI: 10.1186/s13059-025-03501-0] [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: 03/19/2024] [Accepted: 02/05/2025] [Indexed: 03/03/2025] Open
Abstract
Gene context-essentiality assessment supports precision oncology opportunities. The variability of gene effects inference from loss-of-function screenings across models and technologies limits identifying robust hits. We propose a computational framework named PRODE that integrates gene effects with protein-protein interactions to generate neighborhood-informed essential (NIE) and neighborhood-informed context essential (NICE) scores. It outperforms the canonical gene effect approach in recovering missed essential genes in shRNA screens and prioritizing context-essential hits from CRISPR-KO screens, as supported by in vitro validations. Applied to Her2 + breast cancer tumor samples, PRODE identifies oxidative phosphorylation genes as vulnerabilities with prognostic value, highlighting new therapeutic opportunities.
Collapse
Affiliation(s)
- Thomas Cantore
- Laboratory of Computational and Functional Oncology, Department of Cellular, Computational, and Integrative Biology, University of Trento, Via Sommarive 9, Trento, 38123, Italy
| | - Paola Gasperini
- Laboratory of Computational and Functional Oncology, Department of Cellular, Computational, and Integrative Biology, University of Trento, Via Sommarive 9, Trento, 38123, Italy
| | - Riccardo Bevilacqua
- Laboratory of Computational and Functional Oncology, Department of Cellular, Computational, and Integrative Biology, University of Trento, Via Sommarive 9, Trento, 38123, Italy
| | - Yari Ciani
- Laboratory of Computational and Functional Oncology, Department of Cellular, Computational, and Integrative Biology, University of Trento, Via Sommarive 9, Trento, 38123, Italy
| | - Sanju Sinha
- Cancer Data Science Laboratory (CDSL), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
- Currently at Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory (CDSL), Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Francesca Demichelis
- Laboratory of Computational and Functional Oncology, Department of Cellular, Computational, and Integrative Biology, University of Trento, Via Sommarive 9, Trento, 38123, Italy.
| |
Collapse
|
10
|
Coria AR, Shah A, Shafieinouri M, Taylor SJ, Orgebin E, Guiblet W, Miller JT, Sharma IM, Wu CCC. The integrated stress response regulates 18S nonfunctional rRNA decay in mammals. Mol Cell 2025; 85:787-801.e8. [PMID: 39947182 PMCID: PMC11845294 DOI: 10.1016/j.molcel.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 10/08/2024] [Accepted: 01/15/2025] [Indexed: 02/19/2025]
Abstract
18S nonfunctional rRNA decay (NRD) detects and eliminates translationally nonfunctional 18S rRNA. Although this process is critical for ribosome quality control, the mechanisms underlying nonfunctional 18S rRNA turnover remain elusive, particularly in mammals. Here, we show that mammalian 18S NRD initiates through the integrated stress response (ISR) via GCN2. Nonfunctional 18S rRNA induces translational arrest at start sites. Biochemical analyses demonstrate that ISR activation limits translation initiation and attenuates collisions between scanning 43S preinitiation complexes and stalled nonfunctional ribosomes. The ISR promotes 18S NRD and 40S ribosomal protein turnover by RNF10-mediated ubiquitination. Ultimately, RIOK3 binds the resulting ubiquitinated 40S subunits and facilitates 18S rRNA decay. Overall, mammalian 18S NRD acts through GCN2, followed by ubiquitin-dependent 18S rRNA degradation involving the ubiquitin E3 ligase RNF10 and the atypical protein kinase RIOK3. These findings establish a dynamic feedback mechanism by which the GCN2-RNF10-RIOK3 axis surveils ribosome functionality at the translation initiation step.
Collapse
MESH Headings
- RNA, Ribosomal, 18S/metabolism
- RNA, Ribosomal, 18S/genetics
- Humans
- Animals
- RNA Stability
- Stress, Physiological
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitin-Protein Ligases/genetics
- Ubiquitination
- Protein Serine-Threonine Kinases/metabolism
- Protein Serine-Threonine Kinases/genetics
- HEK293 Cells
- Ribosome Subunits, Small, Eukaryotic/metabolism
- Ribosome Subunits, Small, Eukaryotic/genetics
- Mice
- Peptide Chain Initiation, Translational
- Protein Biosynthesis
- Ribosomal Proteins/metabolism
- Ribosomal Proteins/genetics
- HeLa Cells
Collapse
Affiliation(s)
- Aaztli R Coria
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Akruti Shah
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Mohammad Shafieinouri
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Sarah J Taylor
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Emilien Orgebin
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Wilfried Guiblet
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Jennifer T Miller
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Indra Mani Sharma
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | - Colin Chih-Chien Wu
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA.
| |
Collapse
|
11
|
Johri P, Charlesworth B. A gene-based model of fitness and its implications for genetic variation: linkage disequilibrium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.12.612686. [PMID: 40027714 PMCID: PMC11870398 DOI: 10.1101/2024.09.12.612686] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
A widely used model of the effects of mutations on fitness (the "sites" model) assumes that heterozygous recessive or partially recessive deleterious mutations at different sites in a gene complement each other, similarly to mutations in different genes. However, the general lack of complementation between major effect allelic mutations suggests an alternative possibility, which we term the "gene" model. This assumes that a pair of heterozygous deleterious mutations in trans behave effectively as homozygotes, so that the fitnesses of trans heterozygotes are lower than those of cis heterozygotes. We examine the properties of the two different models, using both analytical and simulation methods. We show that the gene model predicts positive linkage disequilibrium (LD) between deleterious variants within the coding sequence, under conditions when the sites model predicts zero or slightly negative LD. We also show that focussing on rare variants when examining patterns of LD, especially with Lewontin's D ' measure, is likely to produce misleading results with respect to inferences concerning the causes of the sign of LD. Synergistic epistasis between pairs of mutations was also modeled; it is less likely to produce negative LD under the gene model than the sites model. The theoretical results are discussed in relation to patterns of LD in natural populations of several species.
Collapse
|
12
|
Chitra U, Arnold B, Raphael BJ. Resolving discrepancies between chimeric and multiplicative measures of higher-order epistasis. Nat Commun 2025; 16:1711. [PMID: 39962081 PMCID: PMC11833126 DOI: 10.1038/s41467-025-56986-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 02/06/2025] [Indexed: 02/20/2025] Open
Abstract
Epistasis - the interaction between alleles at different genetic loci - plays a fundamental role in biology. However, several recent approaches quantify epistasis using a chimeric formula that measures deviations from a multiplicative fitness model on an additive scale, thus mixing two scales. Here, we show that for pairwise interactions, the chimeric formula yields a different magnitude but the same sign of epistasis compared to the multiplicative formula that measures both fitness and deviations on a multiplicative scale. However, for higher-order interactions, we show that the chimeric formula can have both different magnitude and sign compared to the multiplicative formula. We resolve these inconsistencies by deriving mathematical relationships between the different epistasis formulae and different parametrizations of the multivariate Bernoulli distribution. We argue that the chimeric formula does not appropriately model interactions between the Bernoulli random variables. In simulations, we show that the chimeric formula is less accurate than the classical multiplicative/additive epistasis formulae and may falsely detect higher-order epistasis. Analyzing multi-gene knockouts in yeast, multi-way drug interactions in E. coli, and deep mutational scanning of several proteins, we find that approximately 10% to 60% of inferred higher-order interactions change sign using the multiplicative/additive formula compared to the chimeric formula.
Collapse
Affiliation(s)
- Uthsav Chitra
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Brian Arnold
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
13
|
Dibyachintan S, Dubé AK, Bradley D, Lemieux P, Dionne U, Landry CR. Cryptic genetic variation shapes the fate of gene duplicates in a protein interaction network. Nat Commun 2025; 16:1530. [PMID: 39934115 PMCID: PMC11814230 DOI: 10.1038/s41467-025-56597-0] [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: 04/16/2024] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Paralogous genes are often functionally redundant for long periods of time. While their functions are preserved, paralogs accumulate cryptic changes in sequence and expression, which could modulate the impact of future mutations through epistasis. We examine the impact of mutations on redundant myosin proteins that have maintained the same binding preference despite having accumulated differences in expression levels and amino acid substitutions in the last 100 million years. By quantifying the impact of all single-amino acid substitutions in their SH3 domains on the physical interaction with their interaction partners, we show that the same mutations in the paralogous SH3s change binding in a paralog-specific and interaction partner-specific manner. This contingency is explained by the difference in promoter strength of the two paralogous myosin genes and epistatic interactions between the mutations introduced and cryptic divergent sites within the SH3s. One significant consequence of this contingency is that while some mutations would be sufficient to nonfunctionalize one paralog, they would have minimal impact on the other. Our results reveal how cryptic divergence, which accumulates while maintaining functional redundancy in cellular networks, could bias gene duplicates to specific fates.
Collapse
Affiliation(s)
- Soham Dibyachintan
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l'Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Centre de Recherche en Données Massives de l'Université Laval, Université Laval, Québec, QC, Canada
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Université Laval, Québec, QC, Canada
| | - Alexandre K Dubé
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l'Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Centre de Recherche en Données Massives de l'Université Laval, Université Laval, Québec, QC, Canada
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Université Laval, Québec, QC, Canada
- Département de Biologie, Université Laval, Québec, QC, Canada
| | - David Bradley
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l'Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Centre de Recherche en Données Massives de l'Université Laval, Université Laval, Québec, QC, Canada
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Université Laval, Québec, QC, Canada
- Département de Biologie, Université Laval, Québec, QC, Canada
| | - Pascale Lemieux
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l'Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Centre de Recherche en Données Massives de l'Université Laval, Université Laval, Québec, QC, Canada
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Université Laval, Québec, QC, Canada
| | - Ugo Dionne
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l'Ingénierie et les Applications des Protéines, Québec, QC, Canada
- Centre de Recherche en Données Massives de l'Université Laval, Université Laval, Québec, QC, Canada
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Christian R Landry
- PROTEO-Regroupement Québécois de Recherche sur la Fonction, l'Ingénierie et les Applications des Protéines, Québec, QC, Canada.
- Centre de Recherche en Données Massives de l'Université Laval, Université Laval, Québec, QC, Canada.
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada.
- Département de Biochimie, de Microbiologie et de Bio-Informatique, Université Laval, Québec, QC, Canada.
- Département de Biologie, Université Laval, Québec, QC, Canada.
| |
Collapse
|
14
|
Wang Y, Sun Y, Lin B, Zhang H, Luo X, Liu Y, Jin X, Zhu D. SEGT-GO: a graph transformer method based on PPI serialization and explanatory artificial intelligence for protein function prediction. BMC Bioinformatics 2025; 26:46. [PMID: 39930351 PMCID: PMC11808960 DOI: 10.1186/s12859-025-06059-7] [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: 11/05/2024] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND A massive amount of protein sequences have been obtained, but their functions remain challenging to discern. In recent research on protein function prediction, Protein-Protein Interaction (PPI) Networks have played a crucial role. Uncovering potential function relationships between distant proteins within PPI networks is essential for improving the accuracy of protein function prediction. Most current studies attempt to capture these distant relationships by stacking graph network layers, but performance gains diminish as the number of layers increases. RESULTS To further explore the potential functional relationships between multi-hop proteins in PPI networks, this paper proposes SEGT-GO, a Graph Transformer method based on PPI multi-hop neighborhood Serialization and Explainable artificial intelligence for large-scale multispecies protein function prediction. The multi-hop neighborhood serialization maps multi-hop information in the PPI Network into serialized feature embeddings, enabling the Graph Transformer to learn deeper functional features within the PPI Network. Based on game theory, the SHAP eXplainable Artificial Intelligence (XAI) framework optimizes model input and filters out feature noise, enhancing model performance. CONCLUSIONS Compared to the advanced network method DeepGraphGO, SEGT-GO achieves more competitive results in standard large-scale datasets and superior results on small ones, validating its ability to extract functional information from deep proteins. Furthermore, SEGT-GO achieves superior results in cross-species learning and prediction of the functions of unseen proteins, further proving the method's strong generalization.
Collapse
Affiliation(s)
- Yansong Wang
- School of Computer Science and Technology, Harbin Institute of Technology Weihai Campus, Weihai, 264209, China
| | - Yundong Sun
- School of Computer Science and Technology, Harbin Institute of Technology Weihai Campus, Weihai, 264209, China
- Department of Electronic Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Baohui Lin
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, 518118, China
| | - Haotian Zhang
- School of Computer Science and Technology, Harbin Institute of Technology Weihai Campus, Weihai, 264209, China
| | - Xiaoling Luo
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Yumeng Liu
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, 518118, China
| | - Xiaopeng Jin
- College of Big Data and Internet, Shenzhen Technology University, Shenzhen, 518118, China.
| | - Dongjie Zhu
- School of Computer Science and Technology, Harbin Institute of Technology Weihai Campus, Weihai, 264209, China.
| |
Collapse
|
15
|
Pržulj N, Malod-Dognin N. Simplicity within biological complexity. BIOINFORMATICS ADVANCES 2025; 5:vbae164. [PMID: 39927291 PMCID: PMC11805345 DOI: 10.1093/bioadv/vbae164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/01/2024] [Accepted: 10/23/2024] [Indexed: 02/11/2025]
Abstract
Motivation Heterogeneous, interconnected, systems-level, molecular (multi-omic) data have become increasingly available and key in precision medicine. We need to utilize them to better stratify patients into risk groups, discover new biomarkers and targets, repurpose known and discover new drugs to personalize medical treatment. Existing methodologies are limited and a paradigm shift is needed to achieve quantitative and qualitative breakthroughs. Results In this perspective paper, we survey the literature and argue for the development of a comprehensive, general framework for embedding of multi-scale molecular network data that would enable their explainable exploitation in precision medicine in linear time. Network embedding methods (also called graph representation learning) map nodes to points in low-dimensional space, so that proximity in the learned space reflects the network's topology-function relationships. They have recently achieved unprecedented performance on hard problems of utilizing few omic data in various biomedical applications. However, research thus far has been limited to special variants of the problems and data, with the performance depending on the underlying topology-function network biology hypotheses, the biomedical applications, and evaluation metrics. The availability of multi-omic data, modern graph embedding paradigms and compute power call for a creation and training of efficient, explainable and controllable models, having no potentially dangerous, unexpected behaviour, that make a qualitative breakthrough. We propose to develop a general, comprehensive embedding framework for multi-omic network data, from models to efficient and scalable software implementation, and to apply it to biomedical informatics, focusing on precision medicine and personalized drug discovery. It will lead to a paradigm shift in the computational and biomedical understanding of data and diseases that will open up ways to solve some of the major bottlenecks in precision medicine and other domains.
Collapse
Affiliation(s)
- Nataša Pržulj
- Computational Biology Department, Mohamed bin Zayed University of Artificial Intelligence, Abu Dhabi, 00000, United Arabic Emirates
- Barcelona Supercomputing Center, Barcelona 08034, Spain
- Department of Computer Science, University College London, London WC1E6BT, United Kingdom
- ICREA, Pg. Lluís Companys 23, Barcelona 08010, Spain
| | | |
Collapse
|
16
|
Kingma E, Dolsma F, Iñigo de la Cruz L, Laan L. Saturated Transposon Analysis in Yeast as a one-step method to quantify the fitness effects of gene disruptions on a genome-wide scale. PLoS One 2025; 20:e0312437. [PMID: 39913404 PMCID: PMC11801604 DOI: 10.1371/journal.pone.0312437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 10/07/2024] [Indexed: 02/09/2025] Open
Abstract
Transposon insertion site sequencing (TIS) is a powerful tool that has significantly advanced our knowledge of functional genomics. For example, TIS has been used to identify essential genes of Saccharomyces cerevisiae, screen for antibiotic resistance genes in Klebsiella pneumoniae and determine the set of genes required for virulence of Mycobacterium tuberculosis. While providing valuable insights, these applications of TIS focus on (conditional) gene essentiality and neglect possibly interesting but subtle differences in the importance of genes for fitness. Notably, it has been demonstrated that data obtained from TIS experiments can be used for fitness quantification and the construction of genetic interaction maps, but this potential is only sporadically exploited. Here, we present a method to quantify the fitness of gene disruption mutants using data obtained from a TIS screen developed for the yeast Saccharomyces cerevisiae called SATAY. We show that the mean read count per transposon insertion site provides a metric for fitness that is robust across biological and technical replicate experiments. Importantly, the ability to resolve differences between gene disruption mutants with low fitness depends crucially on the inclusion of insertion sites that are not observed in the sequencing data to estimate the mean. While our method provides reproducible results between replicate SATAY datasets, the obtained fitness distribution differs substantially from those obtained using other techniques. It is currently unclear whether these inconsistencies are due to biological or technical differences between the methods. We end with suggestions for modifications of the SATAY procedure that could improve the resolution of the fitness estimates. Our analysis indicates that increasing the sequencing depth does very little to reduce the uncertainty in the estimates, while replacing the PCR amplification with methods that avoid or reduce the number of amplification cycles will likely be most effective in reducing noise.
Collapse
Affiliation(s)
- Enzo Kingma
- Department of Bionanoscience, Kavli Institute, Delft University of Technology, Delft, Zuid-Holland, The Netherlands
| | - Floor Dolsma
- Department of Bionanoscience, Kavli Institute, Delft University of Technology, Delft, Zuid-Holland, The Netherlands
| | - Leila Iñigo de la Cruz
- Department of Bionanoscience, Kavli Institute, Delft University of Technology, Delft, Zuid-Holland, The Netherlands
| | - Liedewij Laan
- Department of Bionanoscience, Kavli Institute, Delft University of Technology, Delft, Zuid-Holland, The Netherlands
| |
Collapse
|
17
|
Hebert JD, Tang YJ, Szamecz M, Andrejka L, Lopez SS, Petrov DA, Boross G, Winslow MM. Combinatorial In Vivo Genome Editing Identifies Widespread Epistasis and an Accessible Fitness Landscape During Lung Tumorigenesis. Mol Biol Evol 2025; 42:msaf023. [PMID: 39907430 PMCID: PMC11824425 DOI: 10.1093/molbev/msaf023] [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: 05/28/2024] [Revised: 11/15/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025] Open
Abstract
Lung adenocarcinoma, the most common subtype of lung cancer, is genomically complex, with tumors containing tens to hundreds of non-synonymous mutations. However, little is understood about how genes interact with each other to enable the evolution of cancer in vivo, largely due to a lack of methods for investigating genetic interactions in a high-throughput and quantitative manner. Here, we employed a novel platform to generate tumors with inactivation of pairs of ten diverse tumor suppressor genes within an autochthonous mouse model of oncogenic KRAS-driven lung cancer. By quantifying the fitness of tumors with every single and double mutant genotype, we show that most tumor suppressor genetic interactions exhibited negative epistasis, with diminishing returns on tumor fitness. In contrast, Apc inactivation showed positive epistasis with the inactivation of several other genes, including synergistic effects on tumor fitness in combination with Lkb1 or Nf1 inactivation. Sign epistasis was extremely rare, suggesting a surprisingly accessible fitness landscape during lung tumorigenesis. These findings expand our understanding of the interactions that drive tumorigenesis in vivo.
Collapse
Affiliation(s)
- Jess D Hebert
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuning J Tang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Márton Szamecz
- Faculty of Informatics, Eötvös Loránd University, Budapest, Hungary
- National Laboratory for Health Security, Centre for Eco-Epidemiology, Budapest, Hungary
- Institute of Evolution, HUN-REN Centre for Ecological Research, Budapest, Hungary
| | - Laura Andrejka
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven S Lopez
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Dmitri A Petrov
- Department of Biology, Stanford University, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Gábor Boross
- National Laboratory for Health Security, Centre for Eco-Epidemiology, Budapest, Hungary
- Institute of Evolution, HUN-REN Centre for Ecological Research, Budapest, Hungary
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
18
|
Fischbach A, Widlund PO, Hao X, Nyström T. mTOR signaling controls protein aggregation during heat stress and cellular aging in a translation- and Hsf1-independent manner. J Biol Chem 2025; 301:108172. [PMID: 39798875 PMCID: PMC11849620 DOI: 10.1016/j.jbc.2025.108172] [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: 06/25/2024] [Revised: 01/01/2025] [Accepted: 01/05/2025] [Indexed: 01/15/2025] Open
Abstract
The mechanistic target of rapamycin (mTOR) signaling pathway appears central to the aging process as genetic or pharmacological inhibition of mTOR extends lifespan in most eukaryotes tested. While the regulation of protein synthesis by mTOR has been studied in great detail, its impact on protein misfolding and aggregation during stress and aging is less explored. In this study, we identified the mTOR signaling pathway and the linked Seh1-associated complex as central nodes of protein aggregation during heat stress and cellular aging, using Saccharomyces cerevisiae as a model organism. Based on a synthetic genetic array screen, we found that reduced mTOR activity, achieved through deletion of TCO89, an mTORC1 subunit, almost completely prevents protein aggregation during heat stress and aging without reducing global translation rates and independently of an Hsf1-dependent stress response. Conversely, increased mTOR activity, achieved through deletion of NPR3, a Seh1-associated complex subunit, exacerbates protein aggregation, but not by overactivating translation. In summary, our work demonstrates that mTOR signaling is a central contributor to age-associated and heat shock-induced protein aggregation, and that this is unlinked to quantitatively discernable effects on translation and Hsf1.
Collapse
Affiliation(s)
- Arthur Fischbach
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg, Sweden.
| | - Per O Widlund
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg, Sweden
| | - Xinxin Hao
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Nyström
- Institute for Biomedicine, Sahlgrenska Academy, Centre for Ageing and Health-AgeCap, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
19
|
Chadwick SR, Stack-Couture S, Berg MD, Di Gregorio S, Lung B, Genereaux J, Moir RD, Brandl CJ, Willis IM, Snapp EL, Lajoie P. TUDCA modulates drug bioavailability to regulate resistance to acute ER stress in Saccharomyces cerevisiae. Mol Biol Cell 2025; 36:ar13. [PMID: 39661468 PMCID: PMC11809307 DOI: 10.1091/mbc.e24-04-0147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/13/2024] Open
Abstract
Cells counter accumulation of misfolded secretory proteins in the endoplasmic reticulum (ER) through activation of the Unfolded Protein Response (UPR). Small molecules termed chemical chaperones can promote protein folding to alleviate ER stress. The bile acid tauroursodeoxycholic acid (TUDCA) has been described as a chemical chaperone. While promising in models of protein folding diseases, TUDCA's mechanism of action remains unclear. Here, we found TUDCA can rescue growth of yeast treated with the ER stressor tunicamycin (Tm), even in the absence of a functional UPR. In contrast, TUDCA failed to rescue growth on other ER stressors. Nor could TUDCA attenuate chronic UPR associated with specific gene deletions or overexpression of a misfolded mutant secretory protein. Neither pretreatment with nor delayed addition of TUDCA conferred protection against Tm. Importantly, attenuation of Tm-induced toxicity required TUDCA's critical micelle forming concentration, suggesting a mechanism where TUDCA directly sequesters drugs. Indeed, in several assays, TUDCA-treated cells closely resembled cells treated with lower doses of Tm. In addition, we found TUDCA can inhibit dyes from labeling intracellular compartments. Thus, our study challenges the model of TUDCA as a chemical chaperone and suggests that TUDCA decreases drug bioavailability, allowing cells to adapt to ER stress.
Collapse
Affiliation(s)
- Sarah R. Chadwick
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Samuel Stack-Couture
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Matthew D. Berg
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Sonja Di Gregorio
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Bryan Lung
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Julie Genereaux
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Robyn D. Moir
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Christopher J. Brandl
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Ian M. Willis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Erik L. Snapp
- Janelia Research Campus of the Howard Hughes Medical Institute, Ashburn, VA 20147
| | - Patrick Lajoie
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
- Children's Health Research Institute, Lawson Health Research Institute, London, Ontario N6C 2V5, Canada
| |
Collapse
|
20
|
Amissah HA, Antwi MH, Amissah TA, Combs SE, Shevtsov M. More than Just Protein Folding: The Epichaperome, Mastermind of the Cancer Cell. Cells 2025; 14:204. [PMID: 39936995 DOI: 10.3390/cells14030204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/13/2025] Open
Abstract
The epichaperome, a dynamic and integrated network of chaperone proteins, extends its roles beyond basic protein folding to protein stabilization and intracellular signal transduction to orchestrating a multitude of cellular processes critical for tumor survival. In this review, we explore the multifaceted roles of the epichaperome, delving into its diverse cellular locations, factors that modulate its formation and function, its liquid-liquid phase separation, and the key signaling and crosstalk pathways it regulates, including cellular metabolism and intracellular signal transduction. We further highlight techniques for isolating and identifying epichaperome networks, pitfalls, and opportunities. Further, we review the profound implications of the epichaperome for cancer treatment and therapy design, underscoring the need for strategic engineering that hinges on a comprehensive insight into the comprehensive structure and workings of the epichaperome across the heterogeneous cell subpopulations in the tumor milieu. By presenting a holistic view of the epichaperome's functions and mechanisms, we aim to underscore its potential as a key target for novel anti-cancer strategies, revealing that the epichaperome is not merely a piece of protein folding machinery but a mastermind that facilitates the malignant phenotype.
Collapse
Affiliation(s)
- Haneef Ahmed Amissah
- Institute of Life Sciences and Biomedicine, Department of Medical Biology and Biotechnology, School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
- Diagnostics Laboratory Department, Trauma and Specialist Hospital, Winneba CE-122-2486, Central Region, Ghana
| | - Maxwell Hubert Antwi
- Department of Medical Laboratory Science, Faculty of Health and Allied Sciences, Koforidua Technical University, Koforidua EN-112-3991, Eastern Region, Ghana
| | - Tawfeek Ahmed Amissah
- Department of Medical Laboratory Science, Faculty of Health and Allied Sciences, Koforidua Technical University, Koforidua EN-112-3991, Eastern Region, Ghana
| | - Stephanie E Combs
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Maxim Shevtsov
- Department of Radiation Oncology, Technische Universität München (TUM), Klinikum Rechts der Isar, 81675 Munich, Germany
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), Saint Petersburg 194064, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg 197341, Russia
| |
Collapse
|
21
|
Kaduhr L, Mayer K, Schaffrath R, Buchner J, Brinkmann U. Diphthamide synthesis is linked to the eEF2-client chaperone machinery. FEBS Lett 2025. [PMID: 39825589 DOI: 10.1002/1873-3468.15095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/26/2024] [Accepted: 12/15/2024] [Indexed: 01/20/2025]
Abstract
The diphthamide modification of eukaryotic translation elongation factor (eEF2) is important for accurate protein synthesis. While the enzymes for diphthamide synthesis are known, coordination of eEF2 synthesis with the diphthamide modification to maintain only modified eEF2 is unknown. Physical and genetic interactions extracted from BioGRID show a connection between diphthamide synthesis enzymes and chaperones in yeast. This includes the Hsp90 co-chaperones Hgh1 and Cpr7. The respective co-chaperone deletion strains contained eEF2 without diphthamide. Notably, strains deficient in other co-chaperones showed no defect in the eEF2-diphthamide modification. Our results demonstrate that diphthamide synthesis involves not only Dph enzymes but also the eEF2-interacting co-chaperones Hgh1 and Cpr7 and may thus require a conformational state of eEF2 which is maintained by specific (co-)chaperones.
Collapse
Affiliation(s)
- Lars Kaduhr
- Department of Microbiology, Kassel University, Germany
| | - Klaus Mayer
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| | | | - Johannes Buchner
- Center for Protein Assemblies (CPA), Department of Bioscience, School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Ulrich Brinkmann
- Roche Pharma Research and Early Development (pRED), Large Molecule Research, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
22
|
Miao Z, Ren Y, Tarabini A, Yang L, Li H, Ye C, Liti G, Fischer G, Li J, Yue JX. ScRAPdb: an integrated pan-omics database for the Saccharomyces cerevisiae reference assembly panel. Nucleic Acids Res 2025; 53:D852-D863. [PMID: 39470715 PMCID: PMC11701598 DOI: 10.1093/nar/gkae955] [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: 08/14/2024] [Revised: 10/05/2024] [Accepted: 10/10/2024] [Indexed: 10/30/2024] Open
Abstract
As a unicellular eukaryote, the budding yeast Saccharomyces cerevisiae strikes a unique balance between biological complexity and experimental tractability, serving as a long-standing classic model for both basic and applied studies. Recently, S. cerevisiae further emerged as a leading system for studying natural diversity of genome evolution and its associated functional implication at population scales. Having high-quality comparative and functional genomics data are critical for such efforts. Here, we exhaustively expanded the telomere-to-telomere (T2T) S. cerevisiae reference assembly panel (ScRAP) that we previously constructed for 142 strains to cover high-quality genome assemblies and annotations of 264 S. cerevisiae strains from diverse geographical and ecological niches and also 33 outgroup strains from all the other Saccharomyces species complex. We created a dedicated online database, ScRAPdb (https://www.evomicslab.org/db/ScRAPdb/), to host this expanded pangenome collection. Furthermore, ScRAPdb also integrates an array of population-scale pan-omics atlases (pantranscriptome, panproteome and panphenome) and extensive data exploration toolkits for intuitive genomics analyses. All curated data and downstream analysis results can be easily downloaded from ScRAPdb. We expect ScRAPdb to become a highly valuable platform for the yeast community and beyond, leading to a pan-omics understanding of the global genetic and phenotypic diversity.
Collapse
Affiliation(s)
- Zepu Miao
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yifan Ren
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Andrea Tarabini
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratory of Computational and Quantitative Biology, 7-9 Quai Saint Bernard, Paris 75005, France
| | - Ludong Yang
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Huihui Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Chang Ye
- Department of Chemistry, University of Chicago, 929 E 57th Street, Chicago, IL 60637, USA
| | - Gianni Liti
- CNRS, INSERM, IRCAN, Université Côte d’Azur, 28 Avenue de Valombrose, Nice 06107, France
| | - Gilles Fischer
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Laboratory of Computational and Quantitative Biology, 7-9 Quai Saint Bernard, Paris 75005, France
| | - Jing Li
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Jia-Xing Yue
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| |
Collapse
|
23
|
Chen L, Gao Y, Hao X, Yang X, Lindström M, Jiang S, Cao X, Liu H, Nyström T, Sunnerhagen P, Liu B. Stress granule formation is regulated by signaling machinery involving Sch9/Ypk1, sphingolipids, and Ubi4. Theranostics 2025; 15:1987-2005. [PMID: 39897563 PMCID: PMC11780528 DOI: 10.7150/thno.98199] [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: 05/07/2024] [Accepted: 11/18/2024] [Indexed: 02/04/2025] Open
Abstract
Rationale: Stress granules (SGs) are membraneless organelles that are formed in response to various stresses. Multiple cellular processes have been reported to be involved in SG formation. However, the signaling cascades that coordinate SG formation remain to be elucidated. Methods: By performing two high-content imaging-based phenomic screens, we identified multiple signaling components that form a possible signal transduction pathway that regulates SG formation. Results: We found that Sch9 and Ypk1 function in an early step of SG formation, leading to a decrease in intermediate long-chain base sphingolipids (LCBs). This further downregulates the polyubiquitin precursor protein Ubi4 through upregulating the deubiquitinase Ubp3. Decreased levels of cellular free ubiquitin may subsequently facilitate Lsm7 phase separation and thus trigger SG formation. Conclusion: The signaling pathway identified in this work, together with its conserved components, provides valuable clues for understanding the mechanisms underlying SG formation and SG-associated human diseases.
Collapse
Affiliation(s)
- Lihua Chen
- Department of Chemistry and Molecular Biology, University of Gothenburg, S-413 90, Göteborg, Sweden
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuan Gao
- Department of Chemistry and Molecular Biology, University of Gothenburg, S-413 90, Göteborg, Sweden
| | - Xinxin Hao
- Department of Chemistry and Molecular Biology, University of Gothenburg, S-413 90, Göteborg, Sweden
| | - Xiaoxue Yang
- Department of Chemistry and Molecular Biology, University of Gothenburg, S-413 90, Göteborg, Sweden
| | - Michelle Lindström
- Department of Chemistry and Molecular Biology, University of Gothenburg, S-413 90, Göteborg, Sweden
| | - Shan Jiang
- Department of Chemistry and Molecular Biology, University of Gothenburg, S-413 90, Göteborg, Sweden
| | - Xiuling Cao
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
| | - Huisheng Liu
- Guangzhou National Laboratory, Guangzhou, Guangdong, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Thomas Nyström
- Department of Chemistry and Molecular Biology, University of Gothenburg, S-413 90, Göteborg, Sweden
| | - Per Sunnerhagen
- Department of Chemistry and Molecular Biology, University of Gothenburg, S-413 90, Göteborg, Sweden
| | - Beidong Liu
- State Key Laboratory of Subtropical Silviculture, School of Forestry and Biotechnology, Zhejiang A&F University, Lin'an, Hangzhou, 311300, China
- EATRIS Center for Large-scale cell-based screening, Department of Chemistry and Molecular Biology, University of Gothenburg, S-413 90, Göteborg, Sweden
| |
Collapse
|
24
|
Maran SR, Leite AB, Alves GG, Bonifácio BS, Alves CE, Moreira POL, Panessa GM, Prado HMDA, Klippel AH, Cussiol JR, Massirer KB, Ferreira TR, Sacks D, Barbiéri CL, da Silva MS, do Monte-Neto RL, Moretti NS. Leishmania mexicana N-Acetyltransferease 10 Is Important for Polysome Formation and Cell Cycle Progression. Mol Microbiol 2025; 123:60-74. [PMID: 39755945 PMCID: PMC11802183 DOI: 10.1111/mmi.15338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/06/2025]
Abstract
Leishmania presents a complex life cycle that involves both invertebrate and vertebrate hosts. By regulating gene expression, protein synthesis, and metabolism, the parasite can adapt to various environmental conditions. This regulation occurs mainly at the post-transcriptional level and may involve epitranscriptomic modifications of RNAs. Recent studies have shown that mRNAs in humans undergo a modification known as N4-acetylcytidine (ac4C) catalyzed by the enzyme N-acetyltransferase (NAT10), impacting mRNAs stability and translation. Here, we characterized the NAT10 homologue of L. mexicana, finding that the enzyme exhibits all the conserved acetyltransferase domains although failed to functionally complement the Kre33 mutant in Saccharomyces cerevisiae. We also discovered that LmexNAT10 is nuclear, and seems essential, as evidenced by unsuccessful attempts to obtain null mutant parasites. Phenotypic characterization of single-knockout parasites revealed that LmexNAT10 affects the multiplication of procyclic forms and the promastigote-amastigote differentiation. Additionally, in vivo infection studies using the invertebrate vector Lutzomyia longipalpis showed a delay in the parasite differentiation into metacyclics. Finally, we observed changes in the cell cycle progression and protein synthesis in the mutant parasites. Together, these results suggest that LmexNAT10 might be important for parasite differentiation, potentially by regulating ac4C levels.
Collapse
Affiliation(s)
- Suellen Rodrigues Maran
- Laboratório de Biologia Molecular de Patógenos (LBMP), Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (Unifesp), Brazil
| | - Ariely Barbosa Leite
- Laboratório de Biologia Molecular de Patógenos (LBMP), Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (Unifesp), Brazil
| | - Gabriela Gomes Alves
- Laboratório de Biologia Molecular de Patógenos (LBMP), Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (Unifesp), Brazil
| | - Bruno Souza Bonifácio
- Laboratório de Biologia Molecular de Patógenos (LBMP), Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (Unifesp), Brazil
| | - Carlos Eduardo Alves
- Laboratório de Biologia Molecular de Patógenos (LBMP), Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (Unifesp), Brazil
| | - Paulo Otávio Lourenço Moreira
- Grupo de Pesquisas em Biotecnologia Aplicada ao Estudo de Patógenos (BAP) – Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Giovanna Marques Panessa
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Heloísa Monteiro do Amaral Prado
- Center for Molecular Biology and Genetic Engineering (CBMEG), Center for Medicinal Chemistry (CQMED), Universidade Estadual de Campinas-UNICAMP, Campinas, Brazil
| | - Angélica Hollunder Klippel
- Center for Molecular Biology and Genetic Engineering (CBMEG), Center for Medicinal Chemistry (CQMED), Universidade Estadual de Campinas-UNICAMP, Campinas, Brazil
| | - José Renato Cussiol
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Katlin Brauer Massirer
- Center for Molecular Biology and Genetic Engineering (CBMEG), Center for Medicinal Chemistry (CQMED), Universidade Estadual de Campinas-UNICAMP, Campinas, Brazil
| | - Tiago Rodrigues Ferreira
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, USA
| | - David Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, NIH, USA
| | - Clara Lúcia Barbiéri
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), Brazil
| | - Marcelo Santos da Silva
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Rubens Lima do Monte-Neto
- Grupo de Pesquisas em Biotecnologia Aplicada ao Estudo de Patógenos (BAP) – Instituto René Rachou, Fundação Oswaldo Cruz, Belo Horizonte, Minas Gerais, Brasil
| | - Nilmar Silvio Moretti
- Laboratório de Biologia Molecular de Patógenos (LBMP), Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal de São Paulo (Unifesp), Brazil
- Departamento de Microbiologia, Imunologia e Parasitologia, Escola Paulista de Medicina, Universidade Federal de São Paulo (Unifesp), Brazil
- Faculty of Veterinay Medicine, University of Montreal, St-Hyacinthe, QC, Canada
- The Research Group on Infectious Diseases in Production Animals (GREMIP), FMV, University of Montreal, St-Hyacinthe, QC, Canada
| |
Collapse
|
25
|
Fong SH, Kuenzi BM, Mattson NM, Lee J, Sanchez K, Bojorquez-Gomez A, Ford K, Munson BP, Licon K, Bergendahl S, Shen JP, Kreisberg JF, Mali P, Hager JH, White MA, Ideker T. A multilineage screen identifies actionable synthetic lethal interactions in human cancers. Nat Genet 2025; 57:154-164. [PMID: 39558023 DOI: 10.1038/s41588-024-01971-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 10/02/2024] [Indexed: 11/20/2024]
Abstract
Cancers are driven by alterations in diverse genes, creating dependencies that can be therapeutically targeted. However, many genetic dependencies have proven inconsistent across tumors. Here we describe SCHEMATIC, a strategy to identify a core network of highly penetrant, actionable genetic interactions. First, fundamental cellular processes are perturbed by systematic combinatorial knockouts across tumor lineages, identifying 1,805 synthetic lethal interactions (95% unreported). Interactions are then analyzed by hierarchical pooling, revealing that half segregate reliably by tissue type or biomarker status (51%) and a substantial minority are penetrant across lineages (34%). Interactions converge on 49 multigene systems, including MAPK signaling and BAF transcriptional regulatory complexes, which become essential on disruption of polymerases. Some 266 interactions translate to robust biomarkers of drug sensitivity, including frequent genetic alterations in the KDM5C/6A histone demethylases, which sensitize to inhibition of TIPARP (PARP7). SCHEMATIC offers a context-aware, data-driven approach to match genetic alterations to targeted therapies.
Collapse
Affiliation(s)
- Samson H Fong
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Brent M Kuenzi
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Nicole M Mattson
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - John Lee
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kyle Sanchez
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ana Bojorquez-Gomez
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kyle Ford
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Brenton P Munson
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Katherine Licon
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Sarah Bergendahl
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - John Paul Shen
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jason F Kreisberg
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Prashant Mali
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | | | | | - Trey Ideker
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
26
|
Bao L, Zhu Z, Ismail A, Zhu B, Anandan V, Whiteley M, Kitten T, Xu P. Experimental evolution of gene essentiality in bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.600122. [PMID: 39071448 PMCID: PMC11275930 DOI: 10.1101/2024.07.16.600122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Essential gene products carry out fundamental cellular activities in interaction with other components. However, the lack of essential gene mutants and appropriate methodologies to link essential gene functions with their partners poses significant challenges. Here, we have generated deletion mutants in 32 genes previously identified as essential, with 23 mutants showing extremely slow growth in the SK36 strain of Streptococcus sanguinis. The 23 genes corresponding to these mutants encode components of diverse pathways, are widely conserved among bacteria, and are essential in many other bacterial species. Whole-genome sequencing of 243 independently evolved populations of these mutants has identified >1000 spontaneous suppressor mutations in experimental evolution. Many of these mutations define new gene and pathway relationships, such as F1Fo-ATPase/V1Vo-ATPase/TrkA1-H1 that were demonstrated across multiple Streptococcus species. Patterns of spontaneous mutations occurring in essential gene mutants differed from those found in wildtype. While gene duplications occurred rarely and appeared most often at later stages of evolution, substitutions, deletions, and insertions were prevalent in evolved populations. These essential gene deletion mutants and spontaneous mutations fixed in the mutant populations during evolution establish a foundation for understanding gene essentiality and the interaction of essential genes in networks.
Collapse
Affiliation(s)
- Liang Bao
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Zan Zhu
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Ahmed Ismail
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Bin Zhu
- Massey Cancer Center, Virginia Commonwealth University, Virginia, USA
| | - Vysakh Anandan
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Marvin Whiteley
- School of Biological Sciences, Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Georgia, USA
| | - Todd Kitten
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| | - Ping Xu
- Department of Oral and Craniofacial Molecular Biology, Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University, Virginia, USA
| |
Collapse
|
27
|
Russo CJ, Husain K, Murugan A. Soft Modes as a Predictive Framework for Low Dimensional Biological Systems across Scales. ARXIV 2024:arXiv:2412.13637v1. [PMID: 39764393 PMCID: PMC11702803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/21/2025]
Abstract
All biological systems are subject to perturbations: due to thermal fluctuations, external environments, or mutations. Yet, while biological systems are composed of thousands of interacting components, recent high-throughput experiments show that their response to perturbations is surprisingly low-dimensional: confined to only a few stereotyped changes out of the many possible. Here, we explore a unifying dynamical systems framework - soft modes - to explain and analyze low-dimensionality in biology, from molecules to eco-systems. We argue that this one framework of soft modes makes non-trivial predictions that generalize classic ideas from developmental biology to disparate systems, namely: phenocopying, dual buffering, and global epistasis. While some of these predictions have been borne out in experiments, we discuss how soft modes allow for a surprisingly far-reaching and unifying framework in which to analyze data from protein biophysics to microbial ecology.
Collapse
Affiliation(s)
- Christopher Joel Russo
- James Franck Institute, University of Chicago, Chicago, United States
- Program in Biophysical Sciences, University of Chicago, Chicago, United States
| | - Kabir Husain
- James Franck Institute, University of Chicago, Chicago, United States
- Department of Physics, University College London, London, United Kingdom
| | - Arvind Murugan
- James Franck Institute, University of Chicago, Chicago, United States
- Department of Physics, University of Chicago, Chicago, United States
| |
Collapse
|
28
|
Dutcher HA, Gasch AP. Investigating the role of RNA-binding protein Ssd1 in aneuploidy tolerance through network analysis. RNA (NEW YORK, N.Y.) 2024; 31:100-112. [PMID: 39471998 DOI: 10.1261/rna.080199.124] [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: 07/19/2024] [Accepted: 10/15/2024] [Indexed: 11/06/2024]
Abstract
RNA-binding proteins (RBPs) play critical cellular roles by mediating various stages of RNA life cycles. Ssd1, an RBP with pleiotropic effects, has been implicated in aneuploidy tolerance in Saccharomyces cerevisiae but its mechanistic role remains unclear. Here, we used a network-based approach to inform on Ssd1's role in aneuploidy tolerance, by identifying and experimentally perturbing a network of RBPs that share mRNA targets with Ssd1. We identified RBPs whose bound mRNA targets significantly overlap with Ssd1 targets. For 14 identified RBPs, we then used a genetic approach to generate all combinations of genotypes for euploid and aneuploid yeast with an extra copy of chromosome XII, with and without SSD1 and/or the RBP of interest. Deletion of 10 RBPs either exacerbated or alleviated the sensitivity of wild-type and/or ssd1Δ cells to chromosome XII duplication, in several cases indicating genetic interactions with SSD1 in the context of aneuploidy. We integrated these findings with results from a global overexpression screen that identified genes whose duplication complements ssd1Δ aneuploid sensitivity. The resulting network points to a subgroup of proteins with shared roles in translational repression and P-body formation, implicating these functions in aneuploidy tolerance. Our results reveal a role for new RBPs in aneuploidy tolerance and support a model in which Ssd1 mitigates translation-related stresses in aneuploid cells.
Collapse
Affiliation(s)
- H Auguste Dutcher
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| | - Audrey P Gasch
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
- Laboratory of Genetics, University of Wisconsin-Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
29
|
Tran JS, Ward RD, Iruegas-López R, Ebersberger I, Peters JM. Chemical genomics informs antibiotic and essential gene function in Acinetobacter baumannii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.627103. [PMID: 39677645 PMCID: PMC11643038 DOI: 10.1101/2024.12.05.627103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The Gram-negative pathogen, Acinetobacter baumannii , poses a serious threat to human health due to its role in nosocomial infections that are resistant to treatment with current antibiotics. Despite this, our understanding of fundamental A. baumannii biology remains limited, as many essential genes have not been experimentally characterized. These essential genes are critical for bacterial survival and, thus, represent promising targets for drug discovery. Here, we systematically probe the function of essential genes by screening a CRISPR interference knockdown library against a diverse panel of chemical inhibitors, including antibiotics. We find that most essential genes show chemical-gene interactions, allowing insights into both inhibitor and gene function. For instance, knockdown of lipooligosaccharide (LOS) transport genes increased sensitivity to a broad range of chemicals. Cells with defective LOS transport showed cell envelope hyper-permeability that was dependent on continued LOS synthesis. Using phenotypes across our chemical-gene interaction dataset, we constructed an essential gene network linking poorly understood genes to well-characterized genes in cell division and other processes. Finally, our phenotype-structure analysis identified structurally related antibiotics with distinct cellular impacts and suggested potential targets for underexplored inhibitors. This study advances our understanding of essential gene and inhibitor function, providing a valuable resource for mechanistic studies, therapeutic strategies, and future key targets for antibiotic development.
Collapse
|
30
|
Rosas Bringas FR, Yin Z, Yao Y, Boudeman J, Ollivaud S, Chang M. Interstitial telomeric sequences promote gross chromosomal rearrangement via multiple mechanisms. Proc Natl Acad Sci U S A 2024; 121:e2407314121. [PMID: 39602274 PMCID: PMC11626172 DOI: 10.1073/pnas.2407314121] [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: 04/11/2024] [Accepted: 10/10/2024] [Indexed: 11/29/2024] Open
Abstract
Telomeric DNA sequences are difficult to replicate. Replication forks frequently pause or stall at telomeres, which can lead to telomere truncation and dysfunction. In addition to being at chromosome ends, telomere repeats are also present at internal locations within chromosomes, known as interstitial telomeric sequences (ITSs). These sequences are unstable and prone to triggering gross chromosomal rearrangements (GCRs). In this study, we quantitatively examined the effect of ITSs on the GCR rate in Saccharomyces cerevisiae using a genetic assay. We find that the GCR rate increases exponentially with ITS length. This increase can be attributed to the telomere repeat binding protein Rap1 impeding DNA replication and a bias of repairing DNA breaks at or distal to the ITS via de novo telomere addition. Additionally, we performed a genome-wide screen for genes that modulate the rate of ITS-induced GCRs. We find that mutation of core components of the DNA replication machinery leads to an increase in GCRs, but many mutants known to increase the GCR rate in the absence of an ITS do not significantly affect the GCR rate when an ITS is present. We also identified genes that promote the formation of ITS-induced GCRs, including genes with roles in telomere maintenance, nucleotide excision repair, and transcription. Our work thus uncovers multiple mechanisms by which an ITS promotes GCR.
Collapse
Affiliation(s)
- Fernando R. Rosas Bringas
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen9713 AV, The Netherlands
| | - Ziqing Yin
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen9713 AV, The Netherlands
| | - Yue Yao
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen9713 AV, The Netherlands
| | - Jonathan Boudeman
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen9713 AV, The Netherlands
| | - Sandra Ollivaud
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen9713 AV, The Netherlands
| | - Michael Chang
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen9713 AV, The Netherlands
| |
Collapse
|
31
|
Notbohm J, Perica T. Biochemistry and genetics are coming together to improve our understanding of genotype to phenotype relationships. Curr Opin Struct Biol 2024; 89:102952. [PMID: 39522438 DOI: 10.1016/j.sbi.2024.102952] [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: 08/26/2024] [Revised: 10/15/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Since genome sequencing became accessible, determining how specific differences in genotypes lead to complex phenotypes such as disease has become one of the key goals in biomedicine. Predicting effects of sequence variants on cellular or organismal phenotype faces several challenges. First, variants simultaneously affect multiple protein properties and predicting their combined effect is complex. Second, effects of changes in a single protein propagate through the cellular network, which we only partially understand. In this review, we emphasize the importance of both biochemistry and genetics in addressing these challenges. Moreover, we highlight work that blurs the distinction between biochemistry and genetics fields to provide new insights into the genotype-to-phenotype relationships.
Collapse
Affiliation(s)
- Judith Notbohm
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland; Biomolecular Structure and Mechanism PhD Program, Life Science Graduate School Zurich, Switzerland
| | - Tina Perica
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland.
| |
Collapse
|
32
|
Pandita M, Shoket H, Kumar R, Bairwa NK. Genetic Interaction Between F-Box Encoding UCC1 and RRM3 Regulates Growth Rate, Cell Size, and Stress Tolerance in Saccharomyces cerevisiae. J Biochem Mol Toxicol 2024; 38:e70059. [PMID: 39558808 DOI: 10.1002/jbt.70059] [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: 11/16/2023] [Revised: 10/21/2024] [Accepted: 11/05/2024] [Indexed: 11/20/2024]
Abstract
Ucc1, an F-box motif-containing protein of Saccharomyces cerevisiae encoded by UCC1 regulates energy metabolism through proteasomal degradation of citrate synthase Cit2 and inactivation of the glyoxylate cycle when glucose is present as the main carbon source in the growth medium. Rrm3, a Pif1 family DNA helicase, encoded by RRM3 regulates the movement of the replication forks during the DNA replication process. Here in this study, we present evidence of binary genetic interaction between both the genes, UCC1 and RRM3, that determine the growth rate, cell morphology, cell size, apoptosis, and stress response. The absence of both genes UCC1 and RRM3 leads to altered cell morphology, increased growth rate, utilization of alternate carbon sources, resistance to hydrogen peroxide, and susceptibility to acetic acid-induced apoptosis. Further, the genetic interaction network analysis shows both the genes UCC1 and RRM3 interaction through the SGS1 and cross-link among metabolic, glyoxylate, DNA replication, and retrograde signaling pathways.
Collapse
Affiliation(s)
- Monika Pandita
- Genome Stability Regulation Lab, School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu & Kashmir, India
| | - Heena Shoket
- Genome Stability Regulation Lab, School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu & Kashmir, India
| | - Rakesh Kumar
- Cancer Genetics Research Group, School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu & Kashmir, India
| | - Narendra K Bairwa
- Genome Stability Regulation Lab, School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu & Kashmir, India
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu & Kashmir, India
| |
Collapse
|
33
|
Shahmoradi Ghahe S, Drabikowski K, Stasiak M, Topf U. Identification of a Non-canonical Function of Prefoldin Subunit 5 in Proteasome Assembly. J Mol Biol 2024; 436:168838. [PMID: 39490918 DOI: 10.1016/j.jmb.2024.168838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
The prefoldin complex is a heterohexameric, evolutionarily conserved co-chaperone that assists in folding of polypeptides downstream of the protein translation machinery. Loss of prefoldin function leads to impaired solubility of cellular proteins. The degradation of proteins by the proteasome is an integral part of protein homeostasis. Failure of regulated protein degradation can lead to the accumulation of misfolded and defective proteins. We show that prefoldin subunit 5 is required for proteasome activity by contributing to the assembly of the 26S proteasome. In particular, we found that absence of the prefoldin subunit 5 impairs formation of the Rpt ring subcomplex of the proteasome. Concomitant deletion of PFD5 and HSM3, a chaperone for assembly of the ATPase subunits comprising the Rpt ring, exacerbates this effect, suggesting a synergistic relationship between the two factors in proteasome assembly. Thus, our findings reveal a regulatory mechanism wherein prefoldin subunit 5 plays a crucial role in maintaining proteasome integrity, thereby influencing the degradation of proteins.
Collapse
Affiliation(s)
- Somayeh Shahmoradi Ghahe
- Laboratory of Molecular Basis of Aging and Rejuvenation, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| | - Krzysztof Drabikowski
- Laboratory of Biological Chemistry of Metal Ions, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Monika Stasiak
- Laboratory of Molecular Basis of Aging and Rejuvenation, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Ulrike Topf
- Laboratory of Molecular Basis of Aging and Rejuvenation, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
34
|
Yuan L, Sun S, Jiang Y, Zhang Q, Ye L, Zheng CH, Huang DS. scRGCL: a cell type annotation method for single-cell RNA-seq data using residual graph convolutional neural network with contrastive learning. Brief Bioinform 2024; 26:bbae662. [PMID: 39708840 DOI: 10.1093/bib/bbae662] [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: 09/26/2024] [Revised: 11/13/2024] [Accepted: 12/04/2024] [Indexed: 12/23/2024] Open
Abstract
Cell type annotation is a critical step in analyzing single-cell RNA sequencing (scRNA-seq) data. A large number of deep learning (DL)-based methods have been proposed to annotate cell types of scRNA-seq data and have achieved impressive results. However, there are several limitations to these methods. First, they do not fully exploit cell-to-cell differential features. Second, they are developed based on shallow features and lack of flexibility in integrating high-order features in the data. Finally, the low-dimensional gene features may lead to overfitting in neural networks. To overcome those limitations, we propose a novel DL-based model, cell type annotation of single-cell RNA-seq data using residual graph convolutional neural network with contrastive learning (scRGCL), based on residual graph convolutional neural network and contrastive learning for cell type annotation of single-cell RNA-seq data. scRGCL mainly consists of a residual graph convolutional neural network, contrastive learning, and weight freezing. A residual graph convolutional neural network is utilized to extract complex high-order features from data. Contrastive learning can help the model learn meaningful cell-to-cell differential features. Weight freezing can avoid overfitting and help the model discover the impact of specific gene expression on cell type annotation. To verify the effectiveness of scRGCL, we compared its performance with six methods (three shallow learning algorithms and three state-of-the-art DL-based methods) on eight single-cell benchmark datasets from two species (seven in human and one in mouse). Experimental results not only show that scRGCL outperforms competing methods but also demonstrate the generalizability of scRGCL for cell type annotation. scRGCL is available at https://github.com/nathanyl/scRGCL.
Collapse
Affiliation(s)
- Lin Yuan
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Provincial Key Laboratory of Industrial Network and Information System Security, Shandong Fundamental Research Center for Computer Science, 3501 Daxue Road, 250353, Shandong, China
| | - Shengguo Sun
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Provincial Key Laboratory of Industrial Network and Information System Security, Shandong Fundamental Research Center for Computer Science, 3501 Daxue Road, 250353, Shandong, China
| | - Yufeng Jiang
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Provincial Key Laboratory of Industrial Network and Information System Security, Shandong Fundamental Research Center for Computer Science, 3501 Daxue Road, 250353, Shandong, China
| | - Qinhu Zhang
- Ningbo Institute of Digital Twin, Eastern Institute of Technology, 568 Tongxin Road, 315201, Zhejiang, China
| | - Lan Ye
- Cancer Center, The Second Hospital of Shandong University, 247 Beiyuan Street, 250033, Shandong, China
| | - Chun-Hou Zheng
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, School of Artificial Intelligence, Anhui University, 111 Jiulong Road, 230601, Anhui, China
| | - De-Shuang Huang
- Ningbo Institute of Digital Twin, Eastern Institute of Technology, 568 Tongxin Road, 315201, Zhejiang, China
| |
Collapse
|
35
|
Tasnina N, Murali TM. ICoN: integration using co-attention across biological networks. BIOINFORMATICS ADVANCES 2024; 5:vbae182. [PMID: 39801779 PMCID: PMC11723530 DOI: 10.1093/bioadv/vbae182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/24/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025]
Abstract
Motivation Molecular interaction networks are powerful tools for studying cellular functions. Integrating diverse types of networks enhances performance in downstream tasks such as gene module detection and protein function prediction. The challenge lies in extracting meaningful protein feature representations due to varying levels of sparsity and noise across these heterogeneous networks. Results We propose ICoN, a novel unsupervised graph neural network model that takes multiple protein-protein association networks as inputs and generates a feature representation for each protein that integrates the topological information from all the networks. A key contribution of ICoN is exploiting a mechanism called "co-attention" that enables cross-network communication during training. The model also incorporates a denoising training technique, introducing perturbations to each input network and training the model to reconstruct the original network from its corrupted version. Our experimental results demonstrate that ICoN surpasses individual networks across three downstream tasks: gene module detection, gene coannotation prediction, and protein function prediction. Compared to existing unsupervised network integration models, ICoN exhibits superior performance across the majority of downstream tasks and shows enhanced robustness against noise. This work introduces a promising approach for effectively integrating diverse protein-protein association networks, aiming to achieve a biologically meaningful representation of proteins. Availability and implementation The ICoN software is available under the GNU Public License v3 at https://github.com/Murali-group/ICoN.
Collapse
Affiliation(s)
- Nure Tasnina
- Department of Computer Science, Virginia Tech, Blacksburg, VA 24061, United States
| | - T M Murali
- Department of Computer Science, Virginia Tech, Blacksburg, VA 24061, United States
| |
Collapse
|
36
|
Balvert M, Cooper-Knock J, Stamp J, Byrne RP, Mourragui S, van Gils J, Benonisdottir S, Schlüter J, Kenna K, Abeln S, Iacoangeli A, Daub JT, Browning BL, Taş G, Hu J, Wang Y, Alhathli E, Harvey C, Pianesi L, Schulte SC, González-Domínguez J, Garrisson E, Snyder MP, Schönhuth A, Sng LMF, Twine NA. Considerations in the search for epistasis. Genome Biol 2024; 25:296. [PMID: 39563431 PMCID: PMC11574992 DOI: 10.1186/s13059-024-03427-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
Epistasis refers to changes in the effect on phenotype of a unit of genetic information, such as a single nucleotide polymorphism or a gene, dependent on the context of other genetic units. Such interactions are both biologically plausible and good candidates to explain observations which are not fully explained by an additive heritability model. However, the search for epistasis has so far largely failed to recover this missing heritability. We identify key challenges and propose that future works need to leverage idealized systems, known biology and even previously identified epistatic interactions, in order to guide the search for new interactions.
Collapse
Affiliation(s)
| | | | | | - Ross P Byrne
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | | | - Juami van Gils
- Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | | | | | - Sanne Abeln
- Utrecht University, Utrecht, The Netherlands
| | - Alfredo Iacoangeli
- Department of Biostatistics and Health Informatics, King's College London, London, UK
- Department of Basic and Clinical Neuroscience, King's College London, London, UK
- NIHR BRC SLAM NHS Foundation Trust, London, UK
| | | | | | - Gizem Taş
- Tilburg University, Tilburg, The Netherlands
- UMC Utrecht, Utrecht, The Netherlands
| | - Jiajing Hu
- Department of Biostatistics and Health Informatics, King's College London, London, UK
| | - Yan Wang
- UMC Utrecht, Utrecht, The Netherlands
| | | | | | | | - Sara C Schulte
- Algorithmic Bioinformatics and Center for Digital Medicine, Heinrich Heine University, Düsseldorf, Germany
| | | | | | | | | | - Letitia M F Sng
- Commonwealth Scientific and Industrial Research Organisation, Westmead, Australia.
| | - Natalie A Twine
- Commonwealth Scientific and Industrial Research Organisation, Westmead, Australia.
| |
Collapse
|
37
|
Volpiana MW, Nenadic A, Beh CT. Regulation of yeast polarized exocytosis by phosphoinositide lipids. Cell Mol Life Sci 2024; 81:457. [PMID: 39560727 DOI: 10.1007/s00018-024-05483-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/01/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024]
Abstract
Phosphoinositides help steer membrane trafficking routes within eukaryotic cells. In polarized exocytosis, which targets vesicular cargo to sites of polarized growth at the plasma membrane (PM), the two phosphoinositides phosphatidylinositol 4-phosphate (PI4P) and its derivative phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) pave the pathway for vesicle transport from the Golgi to the PM. PI4P is a critical regulator of mechanisms that shape late Golgi membranes for vesicle biogenesis and release. Although enriched in vesicle membranes, PI4P is inexplicably removed from post-Golgi vesicles during their transit to the PM, which drives subsequent steps in exocytosis. At the PM, PI(4,5)P2 recruits effectors that establish polarized membrane sites for targeting the vesicular delivery of secretory cargo. The budding yeast Saccharomyces cerevisiae provides an elegant model to unravel the complexities of phosphoinositide regulation during polarized exocytosis. Here, we review how PI4P and PI(4,5)P2 promote yeast vesicle biogenesis, exocyst complex assembly and vesicle docking at polarized cortical sites, and suggest how these steps might impact related mechanisms of human disease.
Collapse
Affiliation(s)
- Matthew W Volpiana
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Aleksa Nenadic
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada
| | - Christopher T Beh
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada.
- Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
38
|
Buzby C, Plavskin Y, Sartori FM, Tong Q, Vail JK, Siegal ML. Epistasis and cryptic QTL identified using modified bulk segregant analysis of copper resistance in budding yeast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620582. [PMID: 39605464 PMCID: PMC11601411 DOI: 10.1101/2024.10.28.620582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
The contributions of genetic interactions to natural trait variation are challenging to estimate experimentally, as current approaches for detecting epistasis are often underpowered. Powerful mapping approaches such as bulk segregant analysis, wherein individuals with extreme phenotypes are pooled for genotyping, obscure epistasis by averaging over genotype combinations. To accurately characterize and quantify epistasis underlying natural trait variation, we have engineered strains of the budding yeast Saccharomyces cerevisiae to enable crosses where one parent's chromosome is fixed while the rest of the chromosomes segregate. These crosses allow us to use bulk segregant analysis to identify quantitative trait loci (QTL) whose effects depend on alleles on the fixed parental chromosome, indicating a genetic interaction with that chromosome. Our method, which we term epic-QTL (for epistatic-with-chromosome QTL) analysis, can thus identify interaction loci with high statistical power. Here we perform epic-QTL analysis of copper resistance with chromosome I or VIII fixed in a cross between divergent naturally derived strains. We find seven loci that interact significantly with chromosome VIII and none that interact with chromosome I, the smallest of the 16 budding yeast chromosomes. Each of the seven interactions alters the magnitude, rather than the direction, of an additive QTL effect. We also show that fixation of one source of variation - in this case chromosome VIII, which contains the large-effect QTL mapping to CUP1 - increases power to detect the contributions of other loci to trait differences.
Collapse
Affiliation(s)
- Cassandra Buzby
- Department of Biology, New York University, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Yevgeniy Plavskin
- Department of Biology, New York University, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Federica M.O. Sartori
- Department of Biology, New York University, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
- Current affiliation: Department of Oncological Sciences, Mount Sinai, New York, NY, USA
| | - Qiange Tong
- Department of Biology, New York University, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Janessa K. Vail
- Department of Biology, New York University, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Mark L. Siegal
- Department of Biology, New York University, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| |
Collapse
|
39
|
Liu W, Huang Y, Xu Y, Gao X, Zhao Y, Fan S, Geng Y, Zhu S. The combined signatures of programmed cell death and immune landscape provide a prognostic and therapeutic biomarker in the hepatocellular carcinoma. Front Chem 2024; 12:1484310. [PMID: 39600313 PMCID: PMC11591233 DOI: 10.3389/fchem.2024.1484310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks as the fourth most common cause of mortality globally among all cancer types. Programmed cell death (PCD) is a crucial biological mechanism governing cancer progression, tumor expansion, and metastatic dissemination. Furthermore, the tumor microenvironment (TME) is critical in influencing overall survival (OS) and immune responses to immunotherapeutic interventions. From a multi-omics perspective, the combination of PCD and TME could help to predict the survival of HCC patient survival and immunotherapy response. Our study analyzed variations in the PCD- and TME-classifier used in the classification of HCC patients into two subgroups: PCD high-TME low and PCD low-TME high. In the following step, we compared the tumor somatic mutation (TMB), immunotherapy response, and functional annotation of both groups of patients. Lastly, Western Blot (WB) were conducted. The immunohistochemistry (IHC) was performed on the Human Protein Atlas (HPA). In the PCD-TME classifier, 23 PCD-related genes and three immune cell types were identified. Patients' prognoses and responses to therapy could be accurately predicted using this model. The findings of this study provide a new instrument for the clinical management of HCC patients, and they contribute to the development of accurate treatment strategies for these patients.
Collapse
Affiliation(s)
- Wanghu Liu
- Department of General Surgery, Affiliated Hospital of Nantong University, Medicine School of Nantong University, Nantong, China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yang Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Xuanji Gao
- Department of General Surgery, Affiliated Hospital of Nantong University, Medicine School of Nantong University, Nantong, China
| | - Yifan Zhao
- Department of General Surgery, Affiliated Hospital of Nantong University, Medicine School of Nantong University, Nantong, China
| | - Simin Fan
- Department of Nursing, Affiliated Hospital of Nantong University, Nantong, China
| | - Yuanzhi Geng
- Medicine School of Nantong University, Nantong, China
| | - Shajun Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| |
Collapse
|
40
|
Windels SFL, Tello Velasco D, Rotkevich M, Malod-Dognin N, Pržulj N. Graphlet-based hyperbolic embeddings capture evolutionary dynamics in genetic networks. Bioinformatics 2024; 40:btae650. [PMID: 39495120 PMCID: PMC11568109 DOI: 10.1093/bioinformatics/btae650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 09/29/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024] Open
Abstract
MOTIVATION Spatial Analysis of Functional Enrichment (SAFE) is a popular tool for biologists to investigate the functional organization of biological networks via highly intuitive 2D functional maps. To create these maps, SAFE uses Spring embedding to project a given network into a 2D space in which nodes connected in the network are near each other in space. However, many biological networks are scale-free, containing highly connected hub nodes. Because Spring embedding fails to separate hub nodes, it provides uninformative embeddings that resemble a 'hairball'. In addition, Spring embedding only captures direct node connectivity in the network and does not consider higher-order node wiring patterns, which are best captured by graphlets, small, connected, nonisomorphic, induced subgraphs. The scale-free structure of biological networks is hypothesized to stem from an underlying low-dimensional hyperbolic geometry, which novel hyperbolic embedding methods try to uncover. These include coalescent embedding, which projects a network onto a 2D disk. RESULTS To better capture the functional organization of scale-free biological networks, whilst also going beyond simple direct connectivity patterns, we introduce Graphlet Coalescent (GraCoal) embedding, which embeds nodes nearby on a disk if they frequently co-occur on a given graphlet together. We use GraCoal to extend SAFE-based network analysis. Through SAFE-enabled enrichment analysis, we show that GraCoal outperforms graphlet-based Spring embedding in capturing the functional organization of the genetic interaction networks of fruit fly, budding yeast, fission yeast and Escherichia coli. We show that depending on the underlying graphlet, GraCoal embeddings capture different topology-function relationships. We show that triangle-based GraCoal embedding captures functional redundancies between paralogs. AVAILABILITY AND IMPLEMENTATION https://gitlab.bsc.es/swindels/gracoal_embedding.
Collapse
Affiliation(s)
| | - Daniel Tello Velasco
- Barcelona Supercomputing Center, Barcelona 08034, Spain
- Universitat de Barcelona, Barcelona 08007, Spain
| | - Mikhail Rotkevich
- Barcelona Supercomputing Center, Barcelona 08034, Spain
- Universitat Politècnica de Catalunya, Barcelona 08034, Spain
| | | | - Nataša Pržulj
- Barcelona Supercomputing Center, Barcelona 08034, Spain
- ICREA, Barcelona 08010, Spain
- Department of Computer Science, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
41
|
Jansen G, Qi T, Latora V, Amoutzias GD, Delneri D, Oliver SG, Nicosia G. Minimisation of metabolic networks defines a new functional class of genes. Nat Commun 2024; 15:9076. [PMID: 39482321 PMCID: PMC11528065 DOI: 10.1038/s41467-024-52816-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 09/20/2024] [Indexed: 11/03/2024] Open
Abstract
Construction of minimal metabolic networks (MMNs) contributes both to our understanding of the origins of metabolism and to the efficiency of biotechnological processes by preventing the diversion of flux away from product formation. We have designed MMNs using a novel in silico synthetic biology pipeline that removes genes encoding enzymes and transporters from genome-scale metabolic models. The resulting minimal gene-set still ensures both viability and high growth rates. The composition of these MMNs has defined a new functional class of genes termed Network Efficiency Determinants (NEDs). These genes, whilst not essential, are very rarely eliminated in constructing an MMN, suggesting that it is difficult for metabolism to be re-routed to obviate the need for such genes. Moreover, the removal of NED genes from an MMN significantly reduces its global efficiency. Bioinformatic analyses of the NED genes have revealed that not only do these genes have more genetic interactions than the bulk of metabolic genes but their protein products also show more protein-protein interactions. In yeast, NED genes are predominantly single-copy and are highly conserved across evolutionarily distant organisms. These features confirm the importance of the NED genes to the metabolic network, including why they are so rarely excluded during minimisation.
Collapse
Affiliation(s)
- Giorgio Jansen
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Department of Biomedical & Biotechnological Sciences, University of Catania, Catania, Italy
| | - Tanda Qi
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Vito Latora
- School of Mathematical Sciences, Queen Mary University of London, London, UK
- Department of Physics and I.N.F.N., University of Catania, Catania, Italy
| | - Grigoris D Amoutzias
- Bioinformatics Laboratory, Department of Biochemistry & Biotechnology, University of Thessaly, Thessaly, Greece
| | - Daniela Delneri
- Manchester Institute of Biotechnology, University of Manchester, Manchester, UK
| | - Stephen G Oliver
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
| | - Giuseppe Nicosia
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
- Department of Biomedical & Biotechnological Sciences, University of Catania, Catania, Italy.
| |
Collapse
|
42
|
Sen A, Rodriguez-Martinez A, Young-Baird SK, Cox RT. The Drosophila ribonucleoprotein Clueless is required for ribosome biogenesis in vivo. J Biol Chem 2024; 300:107946. [PMID: 39481601 PMCID: PMC11625335 DOI: 10.1016/j.jbc.2024.107946] [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: 04/19/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
As hubs of metabolism, mitochondria contribute critical processes to coordinate and optimize energy and intermediate metabolites. Drosophila Clueless (Clu) and vertebrate CLUH are ribonucleoproteins critical for supporting mitochondrial function; yet do so in multiple ways. Clu-CLUH bind mRNAs, and CLUH regulates mRNA localization and translation of mRNAs encoding proteins destined for mitochondrial import. In addition, Clu associates with ribosomal proteins and translation factors; yet whether it is required for fundamental ribosome function in vivo is not clear. In this study, we examine the Clu interactome and probe Clu's requirement in ribosome biogenesis. We previously showed that Clu associates with ribosomal proteins. In this study, we extend these observations to show that clu null mutants display a significant decrease in overall protein synthesis. In addition, Clu associates with ribosomal proteins in an mRNA-independent manner, suggesting Clu's core ribosomal function may be separate from its role in localizing and translating specific mRNAs. We find that Clu is present in the nucleus and associates with the rRNA processing protein fibrillarin but, surprisingly, that processed rRNA products are normal in the absence of Clu. Furthermore, Clu loss does not affect ribosomal protein levels but does result in a decrease in 40S and 60S ribosomal subunit abundance. Together, these results demonstrate that Clu is present in the nucleus and required for 40S and 60S biogenesis and global translation in vivo. These results highlight the multifaceted role of Clu in supporting cell function through regulation of mRNA encoding mitochondrial proteins and ribosome biogenesis.
Collapse
Affiliation(s)
- Aditya Sen
- Department of Biochemistry and Molecular Biology, Uniformed Services University, Bethesda, Maryland, USA; Henry M. Jackson Foundation, Rockville, Bethesda, USA
| | - Ambar Rodriguez-Martinez
- Department of Biochemistry and Molecular Biology, Uniformed Services University, Bethesda, Maryland, USA; Henry M. Jackson Foundation, Rockville, Bethesda, USA
| | - Sara K Young-Baird
- Department of Biochemistry and Molecular Biology, Uniformed Services University, Bethesda, Maryland, USA
| | - Rachel T Cox
- Department of Biochemistry and Molecular Biology, Uniformed Services University, Bethesda, Maryland, USA.
| |
Collapse
|
43
|
Jakobson CM, Hartl J, Trébulle P, Mülleder M, Jarosz DF, Ralser M. A genome-to-proteome atlas charts natural variants controlling proteome diversity and forecasts their fitness effects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619054. [PMID: 39484408 PMCID: PMC11526991 DOI: 10.1101/2024.10.18.619054] [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: 11/03/2024]
Abstract
Despite abundant genomic and phenotypic data across individuals and environments, the functional impact of most mutations on phenotype remains unclear. Here, we bridge this gap by linking genome to proteome in 800 meiotic progeny from an intercross between two closely related Saccharomyces cerevisiae isolates adapted to distinct niches. Modest genetic distance between the parents generated remarkable proteomic diversity that was amplified in the progeny and captured by 6,476 genotype-protein associations, over 1,600 of which we resolved to single variants. Proteomic adaptation emerged through the combined action of numerous cis- and trans-regulatory mutations, a regulatory architecture that was conserved across the species. Notably, trans-regulatory variants often arose in proteins not traditionally associated with gene regulation, such as enzymes. Moreover, the proteomic consequences of mutations predicted fitness under various stresses. Our study demonstrates that the collective action of natural genetic variants drives dramatic proteome diversification, with molecular consequences that forecast phenotypic outcomes.
Collapse
Affiliation(s)
- Christopher M. Jakobson
- Depasssrtment of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Johannes Hartl
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Biochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Pauline Trébulle
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Michael Mülleder
- Core Facility High-Throughput Mass Spectrometry, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Daniel F. Jarosz
- Depasssrtment of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Markus Ralser
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Biochemistry, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
44
|
Salzler HR, Vandadi V, Sallean JR, Matera AG. Set2 and H3K36 regulate the Drosophila male X chromosome in a context-specific manner, independent from MSL complex spreading. Genetics 2024; 228:iyae168. [PMID: 39417694 PMCID: PMC11631440 DOI: 10.1093/genetics/iyae168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 10/15/2024] [Indexed: 10/19/2024] Open
Abstract
Dosage compensation in Drosophila involves upregulating male X-genes two-fold. This process is carried out by the MSL (male-specific lethal) complex, which binds high-affinity sites and spreads to surrounding genes. Current models of MSL spreading focus on interactions betwen MSL3 (male-specific lethal 3) and Set2-dependent histone marks like trimethylated H3 lysine-36 (H3K36me3). However, Set2 could affect DC via another target, or there could be redundancy between canonical H3.2 and variant H3.3 histones. Furthermore, it is important to parse male-specific effects from those that are X-specific. To discriminate among these possibilities, we employed genomic approaches in H3K36 'residue' and Set2 'writer' mutants. The results confirm a role for Set2 in X-gene regulation, but show that expression trends in males are often mirrored in females. Instead of global, male-specific reduction of X-genes in Set2 or H3K36 mutants, we observe heterogeneous effects. Interestingly, we identified groups of differentially expressed genes (DEGs) whose changes were in opposite directions following loss of H3K36 or Set2, suggesting that H3K36me states have reciprocal functions. In contrast to H4K16R controls, differential expression analysis of combined H3.2K36R/H3.3K36R mutants showed neither consistent reduction in X-gene expression, nor correlation with MSL3 binding. Motif analysis of the DEGs implicated BEAF-32 and other insulator proteins in Set2/H3K36-dependent regulation. Overall, the data are inconsistent with the prevailing model wherein H3K36me3 is essential for spreading the MSL complex to genes along the male X. Rather, we propose that Set2 and H3K36 support DC indirectly, via processes that are utilized by MSL but common to both sexes.
Collapse
Affiliation(s)
- Harmony R Salzler
- Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Vasudha Vandadi
- Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Julia R Sallean
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - A Gregory Matera
- Integrative Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
- RNA Discovery and Lineberger Comprehensive Cancer Centers, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
45
|
Roy S, Adhikary H, Isler S, D'Amours D. The Smc5/6 complex counteracts R-loop formation at highly transcribed genes in cooperation with RNase H2. eLife 2024; 13:e96626. [PMID: 39404251 PMCID: PMC11620742 DOI: 10.7554/elife.96626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 10/07/2024] [Indexed: 12/06/2024] Open
Abstract
The R-loop is a common transcriptional by-product that consists of an RNA-DNA duplex joined to a displaced strand of genomic DNA. While the effects of R-loops on health and disease are well established, there is still an incomplete understanding of the cellular processes responsible for their removal from eukaryotic genomes. Here, we show that a core regulator of chromosome architecture -the Smc5/6 complex- plays a crucial role in the removal of R-loop structures formed during gene transcription. Consistent with this, budding yeast mutants defective in the Smc5/6 complex and enzymes involved in R-loop resolution show strong synthetic interactions and accumulate high levels of RNA-DNA hybrid structures in their chromosomes. Importantly, we demonstrate that the Smc5/6 complex acts on specific types of RNA-DNA hybrid structures in vivo and promotes R-loop degradation by the RNase H2 enzyme in vitro. Collectively, our results reveal a crucial role for the Smc5/6 complex in the removal of toxic R-loops formed at highly transcribed genes and telomeres.
Collapse
Affiliation(s)
- Shamayita Roy
- Ottawa Institute of Systems Biology, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - Hemanta Adhikary
- Ottawa Institute of Systems Biology, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - Sarah Isler
- Ottawa Institute of Systems Biology, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| | - Damien D'Amours
- Ottawa Institute of Systems Biology, Department of Cellular and Molecular Medicine, University of OttawaOttawaCanada
| |
Collapse
|
46
|
Valcárcel LV, San José-Enériz E, Ordoñez R, Apaolaza I, Olaverri-Mendizabal D, Barrena N, Valcárcel A, Garate L, San Miguel J, Pineda-Lucena A, Agirre X, Prósper F, Planes FJ. An automated network-based tool to search for metabolic vulnerabilities in cancer. Nat Commun 2024; 15:8685. [PMID: 39394196 PMCID: PMC11470099 DOI: 10.1038/s41467-024-52725-4] [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: 06/22/2022] [Accepted: 09/18/2024] [Indexed: 10/13/2024] Open
Abstract
The development of computational tools for the systematic prediction of metabolic vulnerabilities of cancer cells constitutes a central question in systems biology. Here, we present gmctool, a freely accessible online tool that allows us to accomplish this task in a simple, efficient and intuitive environment. gmctool exploits the concept of genetic Minimal Cut Sets (gMCSs), a theoretical approach to synthetic lethality based on genome-scale metabolic networks, including a unique database of synthetic lethals computed from Human1, the most recent metabolic reconstruction of human cells. gmctool introduces qualitative and quantitative improvements over our previously developed algorithms to predict, visualize and analyze metabolic vulnerabilities in cancer, demonstrating a superior performance than competing algorithms. A detailed illustration of gmctool is presented for multiple myeloma (MM), an incurable hematological malignancy. We provide in vitro experimental evidence for the essentiality of CTPS1 (CTPS synthase) and UAP1 (UDP-N-Acetylglucosamine Pyrophosphorylase 1) in specific MM patient subgroups.
Collapse
Affiliation(s)
- Luis V Valcárcel
- University of Navarra, Tecnun School of Engineering, Manuel de Lardizábal 13, 20018, San Sebastián, Spain
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Edurne San José-Enériz
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
| | - Raquel Ordoñez
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
| | - Iñigo Apaolaza
- University of Navarra, Tecnun School of Engineering, Manuel de Lardizábal 13, 20018, San Sebastián, Spain
| | - Danel Olaverri-Mendizabal
- University of Navarra, Tecnun School of Engineering, Manuel de Lardizábal 13, 20018, San Sebastián, Spain
| | - Naroa Barrena
- University of Navarra, Tecnun School of Engineering, Manuel de Lardizábal 13, 20018, San Sebastián, Spain
| | - Ana Valcárcel
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
| | - Leire Garate
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
| | - Jesús San Miguel
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
- Departmento de Hematología, Clínica Universidad de Navarra and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain
| | - Antonio Pineda-Lucena
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
| | - Xabier Agirre
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain
| | - Felipe Prósper
- Hemato-Oncology Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, IDISNA, CCUN, Avenida Pío XII 55, 31008, Pamplona, Spain.
- CIBERONC Centro de Investigación Biomédica en Red de Cáncer, 28029, Madrid, Spain.
- Departmento de Hematología, Clínica Universidad de Navarra and CCUN, Universidad de Navarra, Avenida Pío XII 36, 31008, Pamplona, Spain.
| | - Francisco J Planes
- University of Navarra, Tecnun School of Engineering, Manuel de Lardizábal 13, 20018, San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, 31008, Pamplona, Navarra, Spain.
- University of Navarra, Instituto de Ciencia de los Datos e Inteligencia Artificial (DATAI), Campus Universitario, 31008, Pamplona, Spain.
| |
Collapse
|
47
|
Saha E, Fanfani V, Mandros P, Ben Guebila M, Fischer J, Shutta KH, DeMeo DL, Lopes-Ramos CM, Quackenbush J. Bayesian inference of sample-specific coexpression networks. Genome Res 2024; 34:1397-1410. [PMID: 39134413 PMCID: PMC11529861 DOI: 10.1101/gr.279117.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/31/2024] [Indexed: 08/28/2024]
Abstract
Gene regulatory networks (GRNs) are effective tools for inferring complex interactions between molecules that regulate biological processes and hence can provide insights into drivers of biological systems. Inferring coexpression networks is a critical element of GRN inference, as the correlation between expression patterns may indicate that genes are coregulated by common factors. However, methods that estimate coexpression networks generally derive an aggregate network representing the mean regulatory properties of the population and so fail to fully capture population heterogeneity. Bayesian optimized networks obtained by assimilating omic data (BONOBO) is a scalable Bayesian model for deriving individual sample-specific coexpression matrices that recognizes variations in molecular interactions across individuals. For each sample, BONOBO assumes a Gaussian distribution on the log-transformed centered gene expression and a conjugate prior distribution on the sample-specific coexpression matrix constructed from all other samples in the data. Combining the sample-specific gene coexpression with the prior distribution, BONOBO yields a closed-form solution for the posterior distribution of the sample-specific coexpression matrices, thus allowing the analysis of large data sets. We demonstrate BONOBO's utility in several contexts, including analyzing gene regulation in yeast transcription factor knockout studies, the prognostic significance of miRNA-mRNA interaction in human breast cancer subtypes, and sex differences in gene regulation within human thyroid tissue. We find that BONOBO outperforms other methods that have been used for sample-specific coexpression network inference and provides insight into individual differences in the drivers of biological processes.
Collapse
Affiliation(s)
- Enakshi Saha
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Viola Fanfani
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Panagiotis Mandros
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Marouen Ben Guebila
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Jonas Fischer
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Katherine H Shutta
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | - Dawn L DeMeo
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Camila M Lopes-Ramos
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - John Quackenbush
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA;
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| |
Collapse
|
48
|
Togra C, Dhage R, Rajyaguru PI. Tdh3 and Rom2 are functional modulators of a conserved condensate-resident RNA-binding protein, Scd6, in Saccharomyces cerevisiae. Genetics 2024; 228:iyae127. [PMID: 39093296 DOI: 10.1093/genetics/iyae127] [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] [Revised: 06/07/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
Arginine-glycine-glycine motif proteins play a crucial role in determining mRNA fate. Suppressor of clathrin deficiency 6 (Scd6) is a conserved arginine-glycine-glycine motif containing ribonucleoprotein (RNP) condensate-resident, translation repressor, and decapping activator protein in Saccharomyces cerevisiae. Identifying protein factors that can modulate Scd6 function is critical to understanding the regulation of mRNA fate by Scd6. In this study, using an approach that combined mRNA tethering assay with flow cytometry, we screened 50 genes for their role in modulating the translation repression activity of Scd6. We identified 8 conserved modulators with human homologs. Of these, we further characterized in detail guanine nucleotide exchange factor Rho1 multicopy suppressor 2 (Rom2) and glycolytic enzyme triose phosphate dehydrogenase 3 (Tdh3), which, respectively, impede and promote translation repression activity of Scd6. Our study reveals that Rom2 negatively regulates the arginine methylation of Scd6 and antagonizes its localization to P-bodies. Tdh3, on the other hand, promotes Scd6 interaction with Hmt1, thereby promoting the arginine methylation of Scd6 and enhanced eIF4G1 interaction, which is known to promote its repression activity. Identifying these novel modulators provides exciting new insights into the role of a metabolic enzyme of the glycolytic pathway and guanine nucleotide exchange factor implicated in the cell wall integrity pathway in regulating Scd6 function and, thereby, cytoplasmic mRNA fate.
Collapse
Affiliation(s)
- Chitra Togra
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | - Riya Dhage
- Department of Biochemistry, Indian Institute of Science, Bangalore 560012, India
| | | |
Collapse
|
49
|
Ghiaci P, Jouhten P, Martyushenko N, Roca-Mesa H, Vázquez J, Konstantinidis D, Stenberg S, Andrejev S, Grkovska K, Mas A, Beltran G, Almaas E, Patil KR, Warringer J. Highly parallelized laboratory evolution of wine yeasts for enhanced metabolic phenotypes. Mol Syst Biol 2024; 20:1109-1133. [PMID: 39174863 PMCID: PMC11450223 DOI: 10.1038/s44320-024-00059-0] [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: 03/21/2024] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/24/2024] Open
Abstract
Adaptive Laboratory Evolution (ALE) of microorganisms can improve the efficiency of sustainable industrial processes important to the global economy. However, stochasticity and genetic background effects often lead to suboptimal outcomes during laboratory evolution. Here we report an ALE platform to circumvent these shortcomings through parallelized clonal evolution at an unprecedented scale. Using this platform, we evolved 104 yeast populations in parallel from many strains for eight desired wine fermentation-related traits. Expansions of both ALE replicates and lineage numbers broadened the evolutionary search spectrum leading to improved wine yeasts unencumbered by unwanted side effects. At the genomic level, evolutionary gains in metabolic characteristics often coincided with distinct chromosome amplifications and the emergence of side-effect syndromes that were characteristic of each selection niche. Several high-performing ALE strains exhibited desired wine fermentation kinetics when tested in larger liquid cultures, supporting their suitability for application. More broadly, our high-throughput ALE platform opens opportunities for rapid optimization of microbes which otherwise could take many years to accomplish.
Collapse
Affiliation(s)
- Payam Ghiaci
- Department of Chemistry and Molecular Biology, University of Gothenburg, PO Box 462, Gothenburg, 40530, Sweden
- Department of Biorefinery and Energy, High-throughput Centre, Research Institutes of Sweden, Örnsköldsvik, 89250, Sweden
- European Molecular Biology Laboratory, Heidelberg, 69117, Germany
| | - Paula Jouhten
- European Molecular Biology Laboratory, Heidelberg, 69117, Germany
- VTT Technical Research Centre of Finland Ltd, Espoo, 02044 VTT, Finland
- Aalto University, Department of Bioproducts and Biosystems, Espoo, 02150, Finland
| | - Nikolay Martyushenko
- Department of Biotechnology and Food Science, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
| | - Helena Roca-Mesa
- Universitat Rovira i Virgili, Dept. Bioquímica i Biotecnologia, Facultat d'Enologia, Tarragona, 43007, Spain
| | - Jennifer Vázquez
- Universitat Rovira i Virgili, Dept. Bioquímica i Biotecnologia, Facultat d'Enologia, Tarragona, 43007, Spain
- Centro Tecnológico del Vino-VITEC, Carretera de Porrera Km. 1, Falset, 43730, Spain
| | | | - Simon Stenberg
- Department of Chemistry and Molecular Biology, University of Gothenburg, PO Box 462, Gothenburg, 40530, Sweden
| | - Sergej Andrejev
- European Molecular Biology Laboratory, Heidelberg, 69117, Germany
| | | | - Albert Mas
- Universitat Rovira i Virgili, Dept. Bioquímica i Biotecnologia, Facultat d'Enologia, Tarragona, 43007, Spain
| | - Gemma Beltran
- Universitat Rovira i Virgili, Dept. Bioquímica i Biotecnologia, Facultat d'Enologia, Tarragona, 43007, Spain
| | - Eivind Almaas
- Department of Biotechnology and Food Science, NTNU - Norwegian University of Science and Technology, Trondheim, Norway.
| | - Kiran R Patil
- European Molecular Biology Laboratory, Heidelberg, 69117, Germany.
- Medical Research Council (MRC) Toxicology Unit, University of Cambridge, Cambridge, CB2 1QR, UK.
| | - Jonas Warringer
- Department of Chemistry and Molecular Biology, University of Gothenburg, PO Box 462, Gothenburg, 40530, Sweden.
| |
Collapse
|
50
|
Loll-Krippleber R, Jiang YK, Brown GW. pSPObooster: A Plasmid System to Improve Sporulation Efficiency of Saccharomyces cerevisiae Lab Strains. Yeast 2024; 41:585-592. [PMID: 39248173 DOI: 10.1002/yea.3978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/22/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024] Open
Abstract
Common Saccharomyces cerevisiae lab yeast strains derived from S288C have meiotic defects and therefore are poor sporulators. Here, we developed a plasmid system containing corrected alleles of the MKT1 and RME1 genes to rescue the meiotic defects and show that standard BY4741 and BY4742 strains containing the plasmid display faster and more efficient sporulation. The plasmid, pSPObooster, can be maintained as an episome and easily cured or stably integrated into the genome at a single locus. We demonstrate the use of pSPObooster in low- and high-throughput yeast genetic manipulations and show that it can expedite both procedures without impacting strain behavior.
Collapse
Affiliation(s)
- Raphael Loll-Krippleber
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Yangyang Kate Jiang
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Grant W Brown
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|