1
|
Stocksdale JT, Leventhal MJ, Lam S, Xu YX, Wang YO, Wang KQ, Thomas R, Faghihmonzavi Z, Raghav Y, Smith C, Wu J, Miramontes R, Sarda K, Johnston H, Shin MG, Huang T, Foster M, Barch M, Amirani N, Paiz C, Easter L, Duderstadt E, Vaibhav V, Sundararaman N, Felsenfeld DP, Vogt TF, Van Eyk J, Finkbeiner S, Kaye JA, Fraenkel E, Thompson LM. Intersecting impact of CAG repeat and huntingtin knockout in stem cell-derived cortical neurons. Neurobiol Dis 2025; 210:106914. [PMID: 40258535 DOI: 10.1016/j.nbd.2025.106914] [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: 01/18/2025] [Revised: 04/13/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025] Open
Abstract
Huntington's Disease (HD) is caused by a CAG repeat expansion in the gene encoding huntingtin (HTT). While normal HTT function appears impacted by the mutation, the specific pathways unique to CAG repeat expansion versus loss of normal function are unclear. To understand the impact of the CAG repeat expansion, we evaluated biological signatures of HTT knockout (HTT KO) versus those that occur from the CAG repeat expansion by applying multi-omics, live cell imaging, survival analysis and a novel feature-based pipeline to study cortical neurons (eCNs) derived from an isogenic human embryonic stem cell series (RUES2). HTT KO and the CAG repeat expansion influence developmental trajectories of eCNs, with opposing effects on growth. Network analyses of differentially expressed genes and proteins associated with enriched epigenetic motifs identified subnetworks common to CAG repeat expansion and HTT KO that include neuronal differentiation, cell cycle regulation, and mechanisms related to transcriptional repression, and may represent gain-of-function mechanisms that cannot be explained by HTT loss of function alone. A combination of dominant and loss-of-function mechanisms are likely involved in the aberrant neurodevelopmental and neurodegenerative features of HD that can help inform therapeutic strategies.
Collapse
Affiliation(s)
| | - Matthew J Leventhal
- MIT PhD Program in Computational and Systems Biology, Cambridge, MA 02139, USA; MIT Department of Biological Engineering, Cambridge, MA 02139, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stephanie Lam
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Yu-Xin Xu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Yang Oliver Wang
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Keona Q Wang
- Department of Neurobiology and Behavior, UC Irvine, Irvine, CA 92677, USA
| | - Reuben Thomas
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Zohreh Faghihmonzavi
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Yogindra Raghav
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Charlene Smith
- Department of Psychiatry and Human Behavior, UC Irvine, Irvine, CA 92697, USA
| | - Jie Wu
- Department of Biological Chemistry, UC Irvine, Irvine, CA 92697, USA
| | - Ricardo Miramontes
- Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA
| | - Kanchan Sarda
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Heather Johnston
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Min-Gyoung Shin
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Terry Huang
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Mikelle Foster
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Mariya Barch
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Naufa Amirani
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Chris Paiz
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Lindsay Easter
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Erse Duderstadt
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Vineet Vaibhav
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | - Jennifer Van Eyk
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Steve Finkbeiner
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Neurology, University of California San Francisco, San Francisco, CA 94158, USA; Taube/Koret Center for Neurodegenerative Disease Research, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Julia A Kaye
- Center for Systems and Therapeutics, Gladstone Institutes, San Francisco, CA 94158, USA; Taube/Koret Center for Neurodegenerative Disease Research, Gladstone Institutes, San Francisco, CA 94158, USA
| | - Ernest Fraenkel
- MIT PhD Program in Computational and Systems Biology, Cambridge, MA 02139, USA; MIT Department of Biological Engineering, Cambridge, MA 02139, USA
| | - Leslie M Thompson
- Department of Neurobiology and Behavior, UC Irvine, Irvine, CA 92677, USA; Department of Psychiatry and Human Behavior, UC Irvine, Irvine, CA 92697, USA; Department of Biological Chemistry, UC Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, UC Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
2
|
Paysan D, Radhakrishnan A, Zhang X, Shivashankar GV, Uhler C. Image2Reg: Linking chromatin images to gene regulation using genetic and chemical perturbation screens. Cell Syst 2025:101293. [PMID: 40359941 DOI: 10.1016/j.cels.2025.101293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 01/17/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025]
Abstract
Representation learning provides an opportunity to uncover the link between 3D genome organization and gene regulatory networks, thereby connecting the physical and the biochemical space of a cell. Our method, Image2Reg, uses chromatin images obtained in large-scale genetic and chemical perturbation screens. Through convolutional neural networks, Image2Reg generates gene embedding that represents the effect of gene perturbation on chromatin organization. In addition, combining protein-protein interaction data with cell-type-specific transcriptomic data through a graph convolutional network, we obtain a gene embedding that represents the regulatory effect of genes. Finally, Image2Reg learns a map between the resulting physical and biochemical representation of cells, allowing us to predict the perturbed gene modules based on chromatin images. Our results confirm the deep link between chromatin organization and gene regulation and demonstrate that it can be harnessed to identify drug targets and genes upstream of perturbed phenotypes from a simple and inexpensive chromatin staining.
Collapse
Affiliation(s)
- Daniel Paysan
- ETH Zurich, Raemisstrasse 101, Zurich 8092, Switzerland; Paul Scherrer Institute, Forschungsstrasse 111, Villigen 5232, Switzerland
| | - Adityanarayanan Radhakrishnan
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Xinyi Zhang
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - G V Shivashankar
- ETH Zurich, Raemisstrasse 101, Zurich 8092, Switzerland; Paul Scherrer Institute, Forschungsstrasse 111, Villigen 5232, Switzerland.
| | - Caroline Uhler
- Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA.
| |
Collapse
|
3
|
Srivastava R. Advancing precision oncology with AI-powered genomic analysis. Front Pharmacol 2025; 16:1591696. [PMID: 40371349 PMCID: PMC12075946 DOI: 10.3389/fphar.2025.1591696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 04/21/2025] [Indexed: 05/16/2025] Open
Abstract
Multiomics data integration approaches offer a comprehensive functional understanding of biological systems, with significant applications in disease therapeutics. However, the quantitative integration of multiomics data presents a complex challenge, requiring highly specialized computational methods. By providing deep insights into disease-associated molecular mechanisms, multiomics facilitates precision medicine by accounting for individual omics profiles, enabling early disease detection and prevention, aiding biomarker discovery for diagnosis, prognosis, and treatment monitoring, and identifying molecular targets for innovative drug development or the repurposing of existing therapies. AI-driven bioinformatics plays a crucial role in multiomics by computing scores to prioritize available drugs, assisting clinicians in selecting optimal treatments. This review will explain the potential of AI and multiomics data integration for disease understanding and therapeutics. It highlight the challenges in quantitative integration of diverse omics data and clinical workflows involving AI in cancer genomics, addressing the ethical and privacy concerns related to AI-driven applications in oncology. The scope of this text is broad yet focused, providing readers with a comprehensive overview of how AI-powered bioinformatics and integrative multiomics approaches are transforming precision oncology. Understanding bioinformatics in Genomics, it explore the integrative multiomics strategies for drug selection, genome profiling and tumor clonality analysis with clinical application of drug prioritization tools, addressing the technical, ethical, and practical hurdles in deploying AI-driven genomics tools.
Collapse
|
4
|
Kachemov M, Vaibhav V, Smith C, Sundararaman N, Heath M, Pendlebury DF, Matlock A, Lau A, Morozko E, Lim RG, Reidling J, Steffan JS, Van Eyk JE, Thompson LM. Dysregulation of protein SUMOylation networks in Huntington's disease R6/2 mouse striatum. Brain 2025; 148:1212-1227. [PMID: 39391934 PMCID: PMC11969464 DOI: 10.1093/brain/awae319] [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: 05/06/2024] [Revised: 08/13/2024] [Accepted: 09/21/2024] [Indexed: 10/12/2024] Open
Abstract
Huntington's disease is a neurodegenerative disorder caused by an expanded CAG repeat mutation in the Huntingtin (HTT) gene. The mutation impacts neuronal protein homeostasis and cortical/striatal circuitry. SUMOylation is a post-translational modification with broad cellular effects including via modification of synaptic proteins. Here, we used an optimized SUMO protein-enrichment and mass spectrometry method to identify the protein SUMOylation/SUMO interaction proteome in the context of Huntington's disease using R6/2 transgenic and non-transgenic mice. Significant changes in the enrichment of SUMOylated and SUMO-interacting proteins were observed, including those involved in presynaptic function, cytomatrix at the active zone, cytoskeleton organization and glutamatergic signalling. Mitochondrial and RNA-binding proteins also showed altered enrichment. Modified SUMO-associated pathways in Huntington's disease tissue include clathrin-mediated endocytosis signalling, synaptogenesis signalling, synaptic long-term potentiation and SNARE signalling. To evaluate how modulation of SUMOylation might influence functional measures of neuronal activity in Huntington's disease cells in vitro, we used primary neuronal cultures from R6/2 and non-transgenic mice. A receptor internalization assay for the metabotropic glutamate receptor 7 (mGLUR7), a SUMO-enriched protein in the mass spectrometry, showed decreased internalization in R6/2 neurons compared to non-transgenic neurons. SiRNA-mediated knockdown of the E3 SUMO ligase protein inhibitor of activated STAT1 (Pias1), which can SUMO modify mGLUR7, reduced this Huntington's disease phenotype. In addition, microelectrode array analysis of primary neuronal cultures indicated early hyperactivity in Huntington's disease cells, while later time points demonstrated deficits in several measurements of neuronal activity within cortical neurons. Huntington's disease phenotypes were rescued at selected time points following knockdown of Pias1. Collectively, our results provide a mouse brain SUMOome resource and show that significant alterations occur within the post-translational landscape of SUMO-protein interactions of synaptic proteins in Huntington's disease mice, suggesting that targeting of synaptic SUMO networks may provide a proteostatic systems-based therapeutic approach for Huntington's disease and other neurological disorders.
Collapse
Affiliation(s)
- Marketta Kachemov
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Vineet Vaibhav
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Charlene Smith
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92868, USA
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Marie Heath
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Devon F Pendlebury
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Andrea Matlock
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Alice Lau
- Sue and Bill Gross Stem Cell Center, University of California Irvine, Irvine, CA 92697, USA
| | - Eva Morozko
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
| | - Ryan G Lim
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Jack Reidling
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - Joan S Steffan
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92868, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Leslie M Thompson
- Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697, USA
- Department of Psychiatry and Human Behavior, University of California Irvine, Irvine, CA 92868, USA
- Sue and Bill Gross Stem Cell Center, University of California Irvine, Irvine, CA 92697, USA
- Institute of Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
5
|
Liu P, Page D, Ahlquist P, Ong IM, Gitter A. MPAC: a computational framework for inferring pathway activities from multi-omic data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.15.599113. [PMID: 38948762 PMCID: PMC11212914 DOI: 10.1101/2024.06.15.599113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Fully capturing cellular state requires examining genomic, epigenomic, transcriptomic, proteomic, and other assays for a biological sample and comprehensive computational modeling to reason with the complex and sometimes conflicting measurements. Modeling these so-called multi-omic data is especially beneficial in disease analysis, where observations across omic data types may reveal unexpected patient groupings and inform clinical outcomes and treatments. We present Multi-omic Pathway Analysis of Cells (MPAC), a computational framework that interprets multi-omic data through prior knowledge from biological pathways. MPAC uses network relationships encoded in pathways using a factor graph to infer consensus activity levels for proteins and associated pathway entities from multi-omic data, runs permutation testing to eliminate spurious activity predictions, and groups biological samples by pathway activities to prioritize proteins with potential clinical relevance. Using DNA copy number alteration and RNA-seq data from head and neck squamous cell carcinoma patients from The Cancer Genome Atlas as an example, we demonstrate that MPAC predicts a patient subgroup related to immune responses not identified by analysis with either input omic data type alone. Key proteins identified via this subgroup have pathway activities related to clinical outcome as well as immune cell compositions. Our MPAC R package, available at https://bioconductor.org/packages/MPAC, enables similar multi-omic analyses on new datasets.
Collapse
Affiliation(s)
- Peng Liu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - David Page
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Paul Ahlquist
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Irene M Ong
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, Wisconsin, United States of America
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
| |
Collapse
|
6
|
Claros MG, Bullones A, Castro AJ, Lima-Cabello E, Viruel MÁ, Suárez MF, Romero-Aranda R, Fernández-Pozo N, Veredas FJ, Belver A, Alché JDD. Multi-Omic Advances in Olive Tree ( Olea europaea subsp. europaea L.) Under Salinity: Stepping Towards 'Smart Oliviculture'. BIOLOGY 2025; 14:287. [PMID: 40136543 PMCID: PMC11939856 DOI: 10.3390/biology14030287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025]
Abstract
Soil salinisation is threatening crop sustainability worldwide, mainly due to anthropogenic climate change. Molecular mechanisms developed to counteract salinity have been intensely studied in model plants. Nevertheless, the economically relevant olive tree (Olea europaea subsp. europaea L.), being highly exposed to soil salinisation, deserves a specific review to extract the recent genomic advances that support the known morphological and biochemical mechanisms that make it a relative salt-tolerant crop. A comprehensive list of 98 olive cultivars classified by salt tolerance is provided, together with the list of available olive tree genomes and genes known to be involved in salt response. Na+ and Cl- exclusion in leaves and retention in roots seem to be the most prominent adaptations, but cell wall thickening and antioxidant changes are also required for a tolerant response. Several post-translational modifications of proteins are emerging as key factors, together with microbiota amendments, making treatments with biostimulants and chemical compounds a promising approach to enable cultivation in already salinised soils. Low and high-throughput transcriptomics and metagenomics results obtained from salt-sensitive and -tolerant cultivars, and the future advantages of engineering specific metacaspases involved in programmed cell death and autophagy pathways to rapidly raise salt-tolerant cultivars or rootstocks are also discussed. The overview of bioinformatic tools focused on olive tree, combined with machine learning approaches for studying plant stress from a multi-omics perspective, indicates that the development of salt-tolerant cultivars or rootstocks adapted to soil salinisation is progressing. This could pave the way for 'smart oliviculture', promoting more productive and sustainable practices under salt stress.
Collapse
Affiliation(s)
- Manuel Gonzalo Claros
- Institute for Mediterranean and Subtropical Horticulture “La Mayora” (IHSM La Mayora-UMA-CSIC), 29010 Malaga, Spain; (A.B.); (M.Á.V.); (R.R.-A.); (N.F.-P.)
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, 29071 Malaga, Spain;
| | - Amanda Bullones
- Institute for Mediterranean and Subtropical Horticulture “La Mayora” (IHSM La Mayora-UMA-CSIC), 29010 Malaga, Spain; (A.B.); (M.Á.V.); (R.R.-A.); (N.F.-P.)
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, 29071 Malaga, Spain;
| | - Antonio Jesús Castro
- Department of Stress, Development and Signaling of Plants, Plant Reproductive Biology and Advanced Microscopy Laboratory (BReMAP), Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (A.J.C.); (E.L.-C.); (A.B.); (J.d.D.A.)
| | - Elena Lima-Cabello
- Department of Stress, Development and Signaling of Plants, Plant Reproductive Biology and Advanced Microscopy Laboratory (BReMAP), Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (A.J.C.); (E.L.-C.); (A.B.); (J.d.D.A.)
| | - María Ángeles Viruel
- Institute for Mediterranean and Subtropical Horticulture “La Mayora” (IHSM La Mayora-UMA-CSIC), 29010 Malaga, Spain; (A.B.); (M.Á.V.); (R.R.-A.); (N.F.-P.)
| | - María Fernanda Suárez
- Department of Molecular Biology and Biochemistry, Universidad de Málaga, 29071 Malaga, Spain;
| | - Remedios Romero-Aranda
- Institute for Mediterranean and Subtropical Horticulture “La Mayora” (IHSM La Mayora-UMA-CSIC), 29010 Malaga, Spain; (A.B.); (M.Á.V.); (R.R.-A.); (N.F.-P.)
| | - Noé Fernández-Pozo
- Institute for Mediterranean and Subtropical Horticulture “La Mayora” (IHSM La Mayora-UMA-CSIC), 29010 Malaga, Spain; (A.B.); (M.Á.V.); (R.R.-A.); (N.F.-P.)
| | - Francisco J. Veredas
- Department of Computer Science and Programming Languages, Universidad de Málaga, 29071 Malaga, Spain;
| | - Andrés Belver
- Department of Stress, Development and Signaling of Plants, Plant Reproductive Biology and Advanced Microscopy Laboratory (BReMAP), Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (A.J.C.); (E.L.-C.); (A.B.); (J.d.D.A.)
| | - Juan de Dios Alché
- Department of Stress, Development and Signaling of Plants, Plant Reproductive Biology and Advanced Microscopy Laboratory (BReMAP), Estación Experimental del Zaidín, CSIC, 18008 Granada, Spain; (A.J.C.); (E.L.-C.); (A.B.); (J.d.D.A.)
- University Institute of Research on Olive Grove and Olive Oils (INUO), Universidad de Jaén, 23071 Jaen, Spain
| |
Collapse
|
7
|
Stocksdale JT, Leventhal MJ, Lam S, Xu YX, Wang YO, Wang KQ, Tomas R, Faghihmonzavi Z, Raghav Y, Smith C, Wu J, Miramontes R, Sarda K, Johnson H, Shin MG, Huang T, Foster M, Barch M, Armani N, Paiz C, Easter L, Duderstadt E, Vaibhav V, Sundararaman N, Felsenfeld DP, Vogt TF, Van Eyk J, Finkbeiner S, Kaye JA, Fraenkel E, Thompson LM. Intersecting impact of CAG repeat and Huntingtin knockout in stem cell-derived cortical neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.24.639958. [PMID: 40060574 PMCID: PMC11888261 DOI: 10.1101/2025.02.24.639958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Huntington's Disease (HD) is caused by a CAG repeat expansion in the gene encoding Huntingtin (HTT ) . While normal HTT function appears impacted by the mutation, the specific pathways unique to CAG repeat expansion versus loss of normal function are unclear. To understand the impact of the CAG repeat expansion, we evaluated biological signatures of HTT knockout ( HTT KO) versus those that occur from the CAG repeat expansion by applying multi-omics, live cell imaging, survival analysis and a novel feature-based pipeline to study cortical neurons (eCNs) derived from an isogenic human embryonic stem cell series (RUES2). HTT KO and the CAG repeat expansion influence developmental trajectories of eCNs, with opposing effects on the growth. Network analyses of differentially expressed genes and proteins associated with enriched epigenetic motifs identified subnetworks common to CAG repeat expansion and HTT KO that include neuronal differentiation, cell cycle regulation, and mechanisms related to transcriptional repression and may represent gain-of-function mechanisms that cannot be explained by HTT loss of function alone. A combination of dominant and loss-of-function mechanisms are likely involved in the aberrant neurodevelopmental and neurodegenerative features of HD that can help inform therapeutic strategies.
Collapse
|
8
|
Kang B, Murphy M, Ng CW, Leventhal MJ, Huynh N, Im E, Danquah S, Housman DE, Nehme R, Farhi SL, Fraenkel E. CellFIE: Integrating Pathway Discovery With Pooled Profiling of Perturbations Uncovers Pathways of Huntington's Disease, Including Genetic Modifiers of Neuronal Development and Morphology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639023. [PMID: 40027702 PMCID: PMC11870572 DOI: 10.1101/2025.02.19.639023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Genomic screens and GWAS are powerful tools for identifying disease-modifying genes, but it is often challenging to understand the pathways by which these genes function. Here, we take an integrated approach that combines network analysis and an imaging-based pooled genetic perturbation study to examine modifiers of Huntington's disease (HD). The computational analysis highlighted several genes in a subnetwork enriched for modifiers of neuronal development and morphology. To test the functional roles of these genes, we developed an experimental pipeline that allows pooled CRISPRi KD of 21 genes in human iPSC-derived neurons followed by optical analysis of genotypes, neuronal arborization, multiplexed pathway activity and morphological fingerprint readout. This approach recovered known genes involved in morphology and confirmed unexpected links from the network between several genetic modifiers of HD and morphology. Our approach overcomes challenges in pooled measurement of neuronal function and health and could be adapted for other phenotypes in HD and other neurological diseases.
Collapse
Affiliation(s)
- Byunguk Kang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Michael Murphy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christopher W. Ng
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew Joseph Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
| | - Nhan Huynh
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Egun Im
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Serwah Danquah
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - David E. Housman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Samouil L. Farhi
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
9
|
Anwar A, Rana S, Pathak P. Artificial intelligence in the management of metabolic disorders: a comprehensive review. J Endocrinol Invest 2025:10.1007/s40618-025-02548-x. [PMID: 39969797 DOI: 10.1007/s40618-025-02548-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/08/2025] [Indexed: 02/20/2025]
Abstract
This review explores the significant role of artificial intelligence (AI) in managing metabolic disorders like diabetes, obesity, metabolic dysfunction-associated fatty liver disease (MAFLD), and thyroid dysfunction. AI applications in this context encompass early diagnosis, personalized treatment plans, risk assessment, prevention, and biomarker discovery for early and accurate disease management. This review also delves into techniques involving machine learning (ML), deep learning (DL), natural language processing (NLP), computer vision, and reinforcement learning associated with AI and their application in metabolic disorders. The following study also enlightens the challenges and ethical considerations associated with AI implementation, such as data privacy, model interpretability, and bias mitigation. We have reviewed various AI-based tools utilized for the diagnosis and management of metabolic disorders, such as Idx, Guardian Connect system, and DreaMed for diabetes. Further, the paper emphasizes the potential of AI to revolutionize the management of metabolic disorders through collaborations among clinicians and AI experts, the integration of AI into clinical practice, and the necessity for long-term validation studies. The references provided in the paper cover a range of studies related to AI, ML, personalized medicine, metabolic disorders, and diagnostic tools in healthcare, including research on disease diagnostics, personalized therapy, chronic disease management, and the application of AI in diabetes care and nutrition.
Collapse
Affiliation(s)
- Aamir Anwar
- Department of Pharmacy, Amity University, Lucknow campus, 226010, Lucknow, Uttar Pradesh, India
| | - Simran Rana
- Department of Pharmacy, Amity University, Lucknow campus, 226010, Lucknow, Uttar Pradesh, India
| | - Priya Pathak
- Department of Pharmacy, Amity University, Lucknow campus, 226010, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
10
|
Boyd SS, Slawson C, Thompson JA. AMEND 2.0: module identification and multi-omic data integration with multiplex-heterogeneous graphs. BMC Bioinformatics 2025; 26:39. [PMID: 39910456 PMCID: PMC11800622 DOI: 10.1186/s12859-025-06063-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: 11/12/2024] [Accepted: 01/22/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Multi-omic studies provide comprehensive insight into biological systems by evaluating cellular changes between normal and pathological conditions at multiple levels of measurement. Biological networks, which represent interactions or associations between biomolecules, have been highly effective in facilitating omic analysis. However, current network-based methods lack generalizability to accommodate multiple data types across a range of diverse experiments. RESULTS We present AMEND 2.0, an updated active module identification method which can analyze multiplex and/or heterogeneous networks integrated with multi-omic data in a highly generalizable framework, in contrast to existing methods, which are mostly appropriate for at most two specific omic types. It is powered by Random Walk with Restart for multiplex-heterogeneous networks, with additional capabilities including degree bias adjustment and biased random walk for multi-objective module identification. AMEND was applied to two real-world multi-omic datasets: renal cell carcinoma data from The cancer genome atlas and an O-GlcNAc Transferase knockout study. Additional analyses investigate the performance of various subroutines of AMEND on tasks of node ranking and degree bias adjustment. CONCLUSIONS While the analysis of multi-omic datasets in a network context is poised to provide deeper understanding of health and disease, new methods are required to fully take advantage of this increasingly complex data. The current study combines several network analysis techniques into a single versatile method for analyzing biological networks with multi-omic data that can be applied in many diverse scenarios. Software is freely available in the R programming language at https://github.com/samboyd0/AMEND .
Collapse
Affiliation(s)
- Samuel S Boyd
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| | - Chad Slawson
- Department of Biochemistry, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- University of Kansas Cancer Center, Kansas City, KS, 66160, USA
- University of Kansas Alzheimer's Disease Research Center, Fairway, KS, 66205, USA
| | - Jeffrey A Thompson
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- University of Kansas Cancer Center, Kansas City, KS, 66160, USA
| |
Collapse
|
11
|
Lawton ML, Inge MM, Blum BC, Smith-Mahoney EL, Bolzan D, Lin W, McConney C, Porter J, Moore J, Youssef A, Tharani Y, Varelas X, Denis GV, Wong WW, Padhorny D, Kozakov D, Siggers T, Wuchty S, Snyder-Cappione J, Emili A. Multiomic profiling of chronically activated CD4+ T cells identifies drivers of exhaustion and metabolic reprogramming. PLoS Biol 2024; 22:e3002943. [PMID: 39689157 DOI: 10.1371/journal.pbio.3002943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/06/2025] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
Repeated antigen exposure leads to T-cell exhaustion, a transcriptionally and epigenetically distinct cellular state marked by loss of effector functions (e.g., cytotoxicity, cytokine production/release), up-regulation of inhibitory receptors (e.g., PD-1), and reduced proliferative capacity. Molecular pathways underlying T-cell exhaustion have been defined for CD8+ cytotoxic T cells, but which factors drive exhaustion in CD4+ T cells, that are also required for an effective immune response against a tumor or infection, remains unclear. Here, we utilize quantitative proteomic, phosphoproteomic, and metabolomic analyses to characterize the molecular basis of the dysfunctional cell state induced by chronic stimulation of CD4+ memory T cells. We identified a dynamic response encompassing both known and novel up-regulated cell surface receptors, as well as dozens of unexpected transcriptional regulators. Integrated causal network analysis of our combined data predicts the histone acetyltransferase p300 as a driver of aspects of this phenotype following chronic stimulation, which we confirmed via targeted small molecule inhibition. While our integrative analysis also revealed large-scale metabolic reprogramming, our independent investigation confirmed a global remodeling away from glycolysis to a dysfunctional fatty acid oxidation-based metabolism coincident with oxidative stress. Overall, these data provide both insights into the mechanistic basis of CD4+ T-cell exhaustion and serve as a valuable resource for future interventional studies aimed at modulating T-cell dysfunction.
Collapse
Affiliation(s)
- Matthew L Lawton
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Melissa M Inge
- Depart of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Benjamin C Blum
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Erika L Smith-Mahoney
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Dante Bolzan
- Department of Computer Science, University of Miami, Miami, Florida, United States of America
| | - Weiwei Lin
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Christina McConney
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Jacob Porter
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Jarrod Moore
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Ahmed Youssef
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Yashasvi Tharani
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Depart of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Xaralabos Varelas
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Gerald V Denis
- Hematology and Medical Oncology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, United States of America
| | - Dzmitry Padhorny
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, United States of America
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, New York, United States of America
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, New York, United States of America
| | - Trevor Siggers
- Depart of Biology, Boston University, Boston, Massachusetts, United States of America
| | - Stefan Wuchty
- Department of Computer Science, University of Miami, Miami, Florida, United States of America
- Miami Institute of Data Science and Computing, Miami, Florida, United States of America
| | - Jennifer Snyder-Cappione
- Department of Microbiology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Andrew Emili
- Center for Network Systems Biology, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Depart of Biology, Boston University, Boston, Massachusetts, United States of America
- Knight Cancer Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| |
Collapse
|
12
|
Mansoor S, Hamid S, Tuan TT, Park JE, Chung YS. Advance computational tools for multiomics data learning. Biotechnol Adv 2024; 77:108447. [PMID: 39251098 DOI: 10.1016/j.biotechadv.2024.108447] [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: 05/19/2024] [Revised: 09/01/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
The burgeoning field of bioinformatics has seen a surge in computational tools tailored for omics data analysis driven by the heterogeneous and high-dimensional nature of omics data. In biomedical and plant science research multi-omics data has become pivotal for predictive analytics in the era of big data necessitating sophisticated computational methodologies. This review explores a diverse array of computational approaches which play crucial role in processing, normalizing, integrating, and analyzing omics data. Notable methods such similarity-based methods, network-based approaches, correlation-based methods, Bayesian methods, fusion-based methods and multivariate techniques among others are discussed in detail, each offering unique functionalities to address the complexities of multi-omics data. Furthermore, this review underscores the significance of computational tools in advancing our understanding of data and their transformative impact on research.
Collapse
Affiliation(s)
- Sheikh Mansoor
- Department of Plant Resources and Environment, Jeju National University, 63243, Republic of Korea
| | - Saira Hamid
- Watson Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Pulwama, J&K, India
| | - Thai Thanh Tuan
- Department of Plant Resources and Environment, Jeju National University, 63243, Republic of Korea; Multimedia Communications Laboratory, University of Information Technology, Ho Chi Minh city 70000, Vietnam; Multimedia Communications Laboratory, Vietnam National University, Ho Chi Minh city 70000, Vietnam
| | - Jong-Eun Park
- Department of Animal Biotechnology, College of Applied Life Science, Jeju National University, Jeju, Jeju-do, Republic of Korea.
| | - Yong Suk Chung
- Department of Plant Resources and Environment, Jeju National University, 63243, Republic of Korea.
| |
Collapse
|
13
|
Hayes CN, Nakahara H, Ono A, Tsuge M, Oka S. From Omics to Multi-Omics: A Review of Advantages and Tradeoffs. Genes (Basel) 2024; 15:1551. [PMID: 39766818 PMCID: PMC11675490 DOI: 10.3390/genes15121551] [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: 11/09/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Bioinformatics is a rapidly evolving field charged with cataloging, disseminating, and analyzing biological data. Bioinformatics started with genomics, but while genomics focuses more narrowly on the genes comprising a genome, bioinformatics now encompasses a much broader range of omics technologies. Overcoming barriers of scale and effort that plagued earlier sequencing methods, bioinformatics adopted an ambitious strategy involving high-throughput and highly automated assays. However, as the list of omics technologies continues to grow, the field of bioinformatics has changed in two fundamental ways. Despite enormous success in expanding our understanding of the biological world, the failure of bulk methods to account for biologically important variability among cells of the same or different type has led to a major shift toward single-cell and spatially resolved omics methods, which attempt to disentangle the conflicting signals contained in heterogeneous samples by examining individual cells or cell clusters. The second major shift has been the attempt to integrate two or more different classes of omics data in a single multimodal analysis to identify patterns that bridge biological layers. For example, unraveling the cause of disease may reveal a metabolite deficiency caused by the failure of an enzyme to be phosphorylated because a gene is not expressed due to aberrant methylation as a result of a rare germline variant. Conclusions: There is a fine line between superficial understanding and analysis paralysis, but like a detective novel, multi-omics increasingly provides the clues we need, if only we are able to see them.
Collapse
Affiliation(s)
- C. Nelson Hayes
- Department of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (A.O.); (M.T.); (S.O.)
| | - Hikaru Nakahara
- Department of Clinical and Molecular Genetics, Hiroshima University, Hiroshima 734-8551, Japan;
| | - Atsushi Ono
- Department of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (A.O.); (M.T.); (S.O.)
| | - Masataka Tsuge
- Department of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (A.O.); (M.T.); (S.O.)
- Liver Center, Hiroshima University, Hiroshima 734-8551, Japan
| | - Shiro Oka
- Department of Gastroenterology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan; (A.O.); (M.T.); (S.O.)
| |
Collapse
|
14
|
Wei L, Aitchison JD, Kaushansky A, Mast FD. Systems-level reconstruction of kinase phosphosignaling networks regulating endothelial barrier integrity using temporal data. NPJ Syst Biol Appl 2024; 10:134. [PMID: 39548089 PMCID: PMC11568298 DOI: 10.1038/s41540-024-00468-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Phosphosignaling networks control cellular processes. We built kinase-mediated regulatory networks elicited by thrombin stimulation of brain endothelial cells using two computational strategies: Temporal Pathway Synthesizer (TPS), which uses phosphoproteomics data as input, and Temporally REsolved KInase Network Generation (TREKING), which uses kinase inhibitor screens. TPS and TREKING predicted overlapping barrier-regulatory kinases connected with unique network topology. Each strategy effectively describes regulatory signaling networks and is broadly applicable across biological systems.
Collapse
Affiliation(s)
- Ling Wei
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA.
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98105, USA
- Department of Biochemistry, University of Washington, Seattle, WA, 98105, USA
| | - Alexis Kaushansky
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA
- Department of Pediatrics, University of Washington, Seattle, WA, 98105, USA
- Department of Global Health, University of Washington, Seattle, WA, 98105, USA
| | - Fred D Mast
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, 98109, USA.
- Department of Pediatrics, University of Washington, Seattle, WA, 98105, USA.
| |
Collapse
|
15
|
Zulfiqar M, Singh V, Steinbeck C, Sorokina M. Review on computer-assisted biosynthetic capacities elucidation to assess metabolic interactions and communication within microbial communities. Crit Rev Microbiol 2024; 50:1053-1092. [PMID: 38270170 DOI: 10.1080/1040841x.2024.2306465] [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: 03/13/2023] [Revised: 11/17/2023] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Microbial communities thrive through interactions and communication, which are challenging to study as most microorganisms are not cultivable. To address this challenge, researchers focus on the extracellular space where communication events occur. Exometabolomics and interactome analysis provide insights into the molecules involved in communication and the dynamics of their interactions. Advances in sequencing technologies and computational methods enable the reconstruction of taxonomic and functional profiles of microbial communities using high-throughput multi-omics data. Network-based approaches, including community flux balance analysis, aim to model molecular interactions within and between communities. Despite these advances, challenges remain in computer-assisted biosynthetic capacities elucidation, requiring continued innovation and collaboration among diverse scientists. This review provides insights into the current state and future directions of computer-assisted biosynthetic capacities elucidation in studying microbial communities.
Collapse
Affiliation(s)
- Mahnoor Zulfiqar
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Vinay Singh
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University, Jena, Germany
| | - Christoph Steinbeck
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University, Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Maria Sorokina
- Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University, Jena, Germany
- Data Science and Artificial Intelligence, Research and Development, Pharmaceuticals, Bayer, Berlin, Germany
| |
Collapse
|
16
|
Wei L, Aitchison JD, Kaushansky A, Mast FD. Systems-level reconstruction of kinase phosphosignaling networks regulating endothelial barrier integrity using temporal data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.01.606198. [PMID: 39149238 PMCID: PMC11326140 DOI: 10.1101/2024.08.01.606198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Phosphosignaling networks control cellular processes. We built kinase-mediated regulatory networks elicited by thrombin stimulation of brain endothelial cells using two computational strategies: Temporal Pathway Synthesizer (TPS), which uses phosphoproetiomics data as input, and Temporally REsolved KInase Network Generation (TREKING), which uses kinase inhibitor screens. TPS and TREKING predicted overlapping barrier-regulatory kinases connected with unique network topology. Each strategy effectively describes regulatory signaling networks and is broadly applicable across biological systems.
Collapse
|
17
|
Leventhal MJ, Zanella CA, Kang B, Peng J, Gritsch D, Liao Z, Bukhari H, Wang T, Pao PC, Danquah S, Benetatos J, Nehme R, Farhi S, Tsai LH, Dong X, Scherzer CR, Feany MB, Fraenkel E. An integrative systems-biology approach defines mechanisms of Alzheimer's disease neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.17.585262. [PMID: 38559190 PMCID: PMC10980014 DOI: 10.1101/2024.03.17.585262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Despite years of intense investigation, the mechanisms underlying neuronal death in Alzheimer's disease, the most common neurodegenerative disorder, remain incompletely understood. To define relevant pathways, we integrated the results of an unbiased, genome-scale forward genetic screen for age-associated neurodegeneration in Drosophila with human and Drosophila Alzheimer's disease-associated multi-omics. We measured proteomics, phosphoproteomics, and metabolomics in Drosophila models of Alzheimer's disease and identified Alzheimer's disease human genetic variants that modify expression in disease-vulnerable neurons. We used a network optimization approach to integrate these data with previously published Alzheimer's disease multi-omic data. We computationally predicted and experimentally demonstrated how HNRNPA2B1 and MEPCE enhance tau-mediated neurotoxicity. Furthermore, we demonstrated that the screen hits CSNK2A1 and NOTCH1 regulate DNA damage in Drosophila and human iPSC-derived neural progenitor cells. Our work identifies candidate pathways that could be targeted to ameliorate neurodegeneration in Alzheimer's disease.
Collapse
Affiliation(s)
- Matthew J Leventhal
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Camila A Zanella
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Byunguk Kang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Jiajie Peng
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - David Gritsch
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Zhixiang Liao
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tao Wang
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Ping-Chieh Pao
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Serwah Danquah
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Joseph Benetatos
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ralda Nehme
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Samouil Farhi
- Spatial Technology Platform, Broad Institute of Harvard and MIT, Cambridge, MA USA
| | - Li-Huei Tsai
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Xianjun Dong
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens R Scherzer
- Precision Neurology Program, Brigham and Women's Hospital and Harvard Medical school, Boston, MA, USA
- APDA Center for Advanced Parkinson's Disease Research, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Present address: Stephen and Denise Adams Center of Yale School of Medicine, CT, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ernest Fraenkel
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Lead contact
| |
Collapse
|
18
|
Zeng M, Yang X, Chen Y, Fan J, Cao L, Wang M, Xiao P, Ling Z, Yin Y, Chen Y. A Network and Pathway Analysis of Genes Associated With Atrial Fibrillation. Cardiovasc Ther 2024; 2024:7054039. [PMID: 39742001 PMCID: PMC11470814 DOI: 10.1155/2024/7054039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 08/09/2024] [Indexed: 01/03/2025] Open
Abstract
Background: Atrial fibrillation (AF) is affected by both environmental and genetic factors. Previous genetic association studies, especially genome-wide association studies, revealed a large group of AF-associated genes. However, little is known about the functions and interactions of these genes. Moreover, established genetic variants of AF contribute modestly to AF variance, implying that numerous additional AF-associated genetic variations need to be identified. Hence, a systematic network and pathway analysis is needed. Methods: We retrieved all AF-associated genes from genetic association studies in various databases and performed integrative analyses including pathway enrichment analysis, pathway crosstalk analysis, network analysis, and microarray meta-analysis. Results: We collected 254 AF-associated genes from genetic association studies in various databases. Pathway enrichment analysis revealed the top biological pathways that were enriched in the AF-associated genes related to cardiac electromechanical activity. Pathway crosstalk analysis showed that numerous neuro-endocrine-immune pathways connected AF with various diseases including cancers, inflammatory diseases, and cardiovascular diseases. Furthermore, an AF-specific subnetwork was constructed with the prize-collecting Steiner forest algorithm based on the AF-associated genes, and 24 novel genes that were potentially associated with AF were inferred by the subnetwork. In the microarray meta-analysis, six of the 24 novel genes (APLP1, CREB1, CREBBP, PRMT1, IRAK1, and PLXND1) were expressed differentially in patients with AF and sinus rhythm. Conclusions: AF is not only an isolated disease with abnormal electrophysiological activity but might also share a common genetic basis and biological process with tumors and inflammatory diseases as well as cardiovascular diseases. Moreover, the six novel genes inferred from network analysis might help detect the missing AF risk loci.
Collapse
Affiliation(s)
- Mengying Zeng
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Xian Yang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | | | - Jinqi Fan
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Li Cao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Menghao Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peilin Xiao
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Zhiyu Ling
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Yuehui Yin
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| | - Yunlin Chen
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Cardiac Electrophysiology, Chongqing, China
- Cardiac Arrhythmia Intervention Center of Chongqing Medical Quality Control Center, Chongqing, China
- Chongqing Atrial Fibrillation Center Alliance, Chongqing, China
| |
Collapse
|
19
|
Vitorino R. Transforming Clinical Research: The Power of High-Throughput Omics Integration. Proteomes 2024; 12:25. [PMID: 39311198 PMCID: PMC11417901 DOI: 10.3390/proteomes12030025] [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: 06/17/2024] [Revised: 08/31/2024] [Accepted: 09/02/2024] [Indexed: 09/26/2024] Open
Abstract
High-throughput omics technologies have dramatically changed biological research, providing unprecedented insights into the complexity of living systems. This review presents a comprehensive examination of the current landscape of high-throughput omics pipelines, covering key technologies, data integration techniques and their diverse applications. It looks at advances in next-generation sequencing, mass spectrometry and microarray platforms and highlights their contribution to data volume and precision. In addition, this review looks at the critical role of bioinformatics tools and statistical methods in managing the large datasets generated by these technologies. By integrating multi-omics data, researchers can gain a holistic understanding of biological systems, leading to the identification of new biomarkers and therapeutic targets, particularly in complex diseases such as cancer. The review also looks at the integration of omics data into electronic health records (EHRs) and the potential for cloud computing and big data analytics to improve data storage, analysis and sharing. Despite significant advances, there are still challenges such as data complexity, technical limitations and ethical issues. Future directions include the development of more sophisticated computational tools and the application of advanced machine learning techniques, which are critical for addressing the complexity and heterogeneity of omics datasets. This review aims to serve as a valuable resource for researchers and practitioners, highlighting the transformative potential of high-throughput omics technologies in advancing personalized medicine and improving clinical outcomes.
Collapse
Affiliation(s)
- Rui Vitorino
- iBiMED, Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
- Department of Surgery and Physiology, Cardiovascular R&D Centre—UnIC@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
20
|
Kuşoğlu A, Örnek D, Dansık A, Uzun C, Nur Özkan S, Sarıca S, Yangın K, Özdinç Ş, Sorhun DT, Solcan N, Doğanalp EC, Arlov Ø, Cunningham K, Karaoğlu IC, Kizilel S, Solaroğlu I, Bulutay P, Fırat P, Erus S, Tanju S, Dilege Ş, Vunjak‐Novakovic G, Tuncbag N, Öztürk E. Extracellular Matrix Sulfation in the Tumor Microenvironment Stimulates Cancer Stemness and Invasiveness. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309966. [PMID: 39083319 PMCID: PMC11423251 DOI: 10.1002/advs.202309966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 07/08/2024] [Indexed: 09/26/2024]
Abstract
Tumor extracellular matrices (ECM) exhibit aberrant changes in composition and mechanics compared to normal tissues. Proteoglycans (PG) are vital regulators of cellular signaling in the ECM with the ability to modulate receptor tyrosine kinase (RTK) activation via their sulfated glycosaminoglycan (sGAG) side chains. However, their role on tumor cell behavior is controversial. Here, it is demonstrated that PGs are heavily expressed in lung adenocarcinoma (LUAD) patients in correlation with invasive phenotype and poor prognosis. A bioengineered human lung tumor model that recapitulates the increase of sGAGs in tumors in an organotypic matrix with independent control of stiffness, viscoelasticity, ligand density, and porosity, is developed. This model reveals that increased sulfation stimulates extensive proliferation, epithelial-mesenchymal transition (EMT), and stemness in cancer cells. The focal adhesion kinase (FAK)-phosphatidylinositol 3-kinase (PI3K) signaling axis is identified as a mediator of sulfation-induced molecular changes in cells upon activation of a distinct set of RTKs within tumor-mimetic hydrogels. The study shows that the transcriptomic landscape of tumor cells in response to increased sulfation resembles native PG-rich patient tumors by employing integrative omics and network modeling approaches.
Collapse
|
21
|
Zitnik M, Li MM, Wells A, Glass K, Morselli Gysi D, Krishnan A, Murali TM, Radivojac P, Roy S, Baudot A, Bozdag S, Chen DZ, Cowen L, Devkota K, Gitter A, Gosline SJC, Gu P, Guzzi PH, Huang H, Jiang M, Kesimoglu ZN, Koyuturk M, Ma J, Pico AR, Pržulj N, Przytycka TM, Raphael BJ, Ritz A, Sharan R, Shen Y, Singh M, Slonim DK, Tong H, Yang XH, Yoon BJ, Yu H, Milenković T. Current and future directions in network biology. BIOINFORMATICS ADVANCES 2024; 4:vbae099. [PMID: 39143982 PMCID: PMC11321866 DOI: 10.1093/bioadv/vbae099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 05/31/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024]
Abstract
Summary Network biology is an interdisciplinary field bridging computational and biological sciences that has proved pivotal in advancing the understanding of cellular functions and diseases across biological systems and scales. Although the field has been around for two decades, it remains nascent. It has witnessed rapid evolution, accompanied by emerging challenges. These stem from various factors, notably the growing complexity and volume of data together with the increased diversity of data types describing different tiers of biological organization. We discuss prevailing research directions in network biology, focusing on molecular/cellular networks but also on other biological network types such as biomedical knowledge graphs, patient similarity networks, brain networks, and social/contact networks relevant to disease spread. In more detail, we highlight areas of inference and comparison of biological networks, multimodal data integration and heterogeneous networks, higher-order network analysis, machine learning on networks, and network-based personalized medicine. Following the overview of recent breakthroughs across these five areas, we offer a perspective on future directions of network biology. Additionally, we discuss scientific communities, educational initiatives, and the importance of fostering diversity within the field. This article establishes a roadmap for an immediate and long-term vision for network biology. Availability and implementation Not applicable.
Collapse
Affiliation(s)
- Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, United States
| | - Michelle M Li
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, United States
| | - Aydin Wells
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
- Lucy Family Institute for Data and Society, University of Notre Dame, Notre Dame, IN 46556, United States
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Kimberly Glass
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Deisy Morselli Gysi
- Channing Division of Network Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, United States
- Department of Statistics, Federal University of Paraná, Curitiba, Paraná 81530-015, Brazil
- Department of Physics, Northeastern University, Boston, MA 02115, United States
| | - Arjun Krishnan
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - T M Murali
- Department of Computer Science, Virginia Tech, Blacksburg, VA 24061, United States
| | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA 02115, United States
| | - Sushmita Roy
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53715, United States
- Wisconsin Institute for Discovery, Madison, WI 53715, United States
| | - Anaïs Baudot
- Aix Marseille Université, INSERM, MMG, Marseille, France
| | - Serdar Bozdag
- Department of Computer Science and Engineering, University of North Texas, Denton, TX 76203, United States
- Department of Mathematics, University of North Texas, Denton, TX 76203, United States
| | - Danny Z Chen
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Lenore Cowen
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| | - Kapil Devkota
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53715, United States
- Morgridge Institute for Research, Madison, WI 53715, United States
| | - Sara J C Gosline
- Biological Sciences Division, Pacific Northwest National Laboratory, Seattle, WA 98109, United States
| | - Pengfei Gu
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Pietro H Guzzi
- Department of Medical and Surgical Sciences, University Magna Graecia of Catanzaro, Catanzaro, 88100, Italy
| | - Heng Huang
- Department of Computer Science, University of Maryland College Park, College Park, MD 20742, United States
| | - Meng Jiang
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
| | - Ziynet Nesibe Kesimoglu
- Department of Computer Science and Engineering, University of North Texas, Denton, TX 76203, United States
- National Center of Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20814, United States
| | - Mehmet Koyuturk
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, OH 44106, United States
| | - Jian Ma
- Ray and Stephanie Lane Computational Biology Department, School of Computer Science, Carnegie Mellon University, Pittsburgh, PA 15213, United States
| | - Alexander R Pico
- Institute of Data Science and Biotechnology, Gladstone Institutes, San Francisco, CA 94158, United States
| | - Nataša Pržulj
- Department of Computer Science, University College London, London, WC1E 6BT, England
- ICREA, Catalan Institution for Research and Advanced Studies, Barcelona, 08010, Spain
- Barcelona Supercomputing Center (BSC), Barcelona, 08034, Spain
| | - Teresa M Przytycka
- National Center of Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20814, United States
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ 08544, United States
| | - Anna Ritz
- Department of Biology, Reed College, Portland, OR 97202, United States
| | - Roded Sharan
- School of Computer Science, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Yang Shen
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, United States
| | - Mona Singh
- Department of Computer Science, Princeton University, Princeton, NJ 08544, United States
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, United States
| | - Donna K Slonim
- Department of Computer Science, Tufts University, Medford, MA 02155, United States
| | - Hanghang Tong
- Department of Computer Science, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States
| | - Xinan Holly Yang
- Department of Pediatrics, University of Chicago, Chicago, IL 60637, United States
| | - Byung-Jun Yoon
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, United States
- Computational Science Initiative, Brookhaven National Laboratory, Upton, NY 11973, United States
| | - Haiyuan Yu
- Department of Computational Biology, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, United States
| | - Tijana Milenković
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, IN 46556, United States
- Lucy Family Institute for Data and Society, University of Notre Dame, Notre Dame, IN 46556, United States
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN 46556, United States
| |
Collapse
|
22
|
Arici MK, Tuncbag N. Unveiling hidden connections in omics data via pyPARAGON: an integrative hybrid approach for disease network construction. Brief Bioinform 2024; 25:bbae399. [PMID: 39163205 PMCID: PMC11334722 DOI: 10.1093/bib/bbae399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/26/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024] Open
Abstract
Network inference or reconstruction algorithms play an integral role in successfully analyzing and identifying causal relationships between omics hits for detecting dysregulated and altered signaling components in various contexts, encompassing disease states and drug perturbations. However, accurate representation of signaling networks and identification of context-specific interactions within sparse omics datasets in complex interactomes pose significant challenges in integrative approaches. To address these challenges, we present pyPARAGON (PAgeRAnk-flux on Graphlet-guided network for multi-Omic data integratioN), a novel tool that combines network propagation with graphlets. pyPARAGON enhances accuracy and minimizes the inclusion of nonspecific interactions in signaling networks by utilizing network rather than relying on pairwise connections among proteins. Through comprehensive evaluations on benchmark signaling pathways, we demonstrate that pyPARAGON outperforms state-of-the-art approaches in node propagation and edge inference. Furthermore, pyPARAGON exhibits promising performance in discovering cancer driver networks. Notably, we demonstrate its utility in network-based stratification of patient tumors by integrating phosphoproteomic data from 105 breast cancer tumors with the interactome and demonstrating tumor-specific signaling pathways. Overall, pyPARAGON is a novel tool for analyzing and integrating multi-omic data in the context of signaling networks. pyPARAGON is available at https://github.com/netlab-ku/pyPARAGON.
Collapse
Affiliation(s)
- Muslum Kaan Arici
- Graduate School of Informatics, Middle East Technical University, Ankara 06800, Turkey
| | - Nurcan Tuncbag
- Chemical and Biological Engineering, College of Engineering, Koc University, Istanbul 34450, Turkey
- School of Medicine, Koc University, Istanbul 34450, Turkey
- Koc University Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul 34450, Turkey
| |
Collapse
|
23
|
Giudice G, Chen H, Koutsandreas T, Petsalaki E. phuEGO: A Network-Based Method to Reconstruct Active Signaling Pathways From Phosphoproteomics Datasets. Mol Cell Proteomics 2024; 23:100771. [PMID: 38642805 PMCID: PMC11134849 DOI: 10.1016/j.mcpro.2024.100771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/08/2024] [Accepted: 04/17/2024] [Indexed: 04/22/2024] Open
Abstract
Signaling networks are critical for virtually all cell functions. Our current knowledge of cell signaling has been summarized in signaling pathway databases, which, while useful, are highly biased toward well-studied processes, and do not capture context specific network wiring or pathway cross-talk. Mass spectrometry-based phosphoproteomics data can provide a more unbiased view of active cell signaling processes in a given context, however, it suffers from low signal-to-noise ratio and poor reproducibility across experiments. While progress in methods to extract active signaling signatures from such data has been made, there are still limitations with respect to balancing bias and interpretability. Here we present phuEGO, which combines up-to-three-layer network propagation with ego network decomposition to provide small networks comprising active functional signaling modules. PhuEGO boosts the signal-to-noise ratio from global phosphoproteomics datasets, enriches the resulting networks for functional phosphosites and allows the improved comparison and integration across datasets. We applied phuEGO to five phosphoproteomics data sets from cell lines collected upon infection with SARS CoV2. PhuEGO was better able to identify common active functions across datasets and to point to a subnetwork enriched for known COVID-19 targets. Overall, phuEGO provides a flexible tool to the community for the improved functional interpretation of global phosphoproteomics datasets.
Collapse
Affiliation(s)
- Girolamo Giudice
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridgeshire, United Kingdom
| | - Haoqi Chen
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridgeshire, United Kingdom
| | - Thodoris Koutsandreas
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridgeshire, United Kingdom
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Cambridgeshire, United Kingdom.
| |
Collapse
|
24
|
Bukhari H, Nithianandam V, Battaglia RA, Cicalo A, Sarkar S, Comjean A, Hu Y, Leventhal MJ, Dong X, Feany MB. Transcriptional programs mediating neuronal toxicity and altered glial-neuronal signaling in a Drosophila knock-in tauopathy model. Genome Res 2024; 34:590-605. [PMID: 38599684 PMCID: PMC11146598 DOI: 10.1101/gr.278576.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 04/04/2024] [Indexed: 04/12/2024]
Abstract
Missense mutations in the gene encoding the microtubule-associated protein TAU (current and approved symbol is MAPT) cause autosomal dominant forms of frontotemporal dementia. Multiple models of frontotemporal dementia based on transgenic expression of human TAU in experimental model organisms, including Drosophila, have been described. These models replicate key features of the human disease but do not faithfully recreate the genetic context of the human disorder. Here we use CRISPR-Cas-mediated gene editing to model frontotemporal dementia caused by the TAU P301L mutation by creating the orthologous mutation, P251L, in the endogenous Drosophila tau gene. Flies heterozygous or homozygous for Tau P251L display age-dependent neurodegeneration, display metabolic defects, and accumulate DNA damage in affected neurons. To understand the molecular events promoting neuronal dysfunction and death in knock-in flies, we performed single-cell RNA sequencing on approximately 130,000 cells from brains of Tau P251L mutant and control flies. We found that expression of disease-associated mutant tau altered gene expression cell autonomously in all neuronal cell types identified. Gene expression was also altered in glial cells, suggestive of non-cell-autonomous regulation. Cell signaling pathways, including glial-neuronal signaling, were broadly dysregulated as were brain region and cell type-specific protein interaction networks and gene regulatory programs. In summary, we present here a genetic model of tauopathy that faithfully recapitulates the genetic context and phenotypic features of the human disease, and use the results of comprehensive single-cell sequencing analysis to outline pathways of neurotoxicity and highlight the potential role of non-cell-autonomous changes in glia.
Collapse
Affiliation(s)
- Hassan Bukhari
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Vanitha Nithianandam
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Rachel A Battaglia
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| | - Anthony Cicalo
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
- Genomics and Bioinformatics Hub, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Souvarish Sarkar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Matthew J Leventhal
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- MIT Ph.D. Program in Computational and Systems Biology, Cambridge, Massachusetts 02139, USA
| | - Xianjun Dong
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
- Genomics and Bioinformatics Hub, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
- Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Mel B Feany
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA;
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland 20815, USA
| |
Collapse
|
25
|
Biswas A, Kumari A, Gaikwad DS, Pandey DK. Revolutionizing Biological Science: The Synergy of Genomics in Health, Bioinformatics, Agriculture, and Artificial Intelligence. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2023; 27:550-569. [PMID: 38100404 DOI: 10.1089/omi.2023.0197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
With climate emergency, COVID-19, and the rise of planetary health scholarship, the binary of human and ecosystem health has been deeply challenged. The interdependence of human and nonhuman animal health is increasingly acknowledged and paving the way for new frontiers in integrative biology. The convergence of genomics in health, bioinformatics, agriculture, and artificial intelligence (AI) has ushered in a new era of possibilities and applications. However, the sheer volume of genomic/multiomics big data generated also presents formidable sociotechnical challenges in extracting meaningful biological, planetary health and ecological insights. Over the past few years, AI-guided bioinformatics has emerged as a powerful tool for managing, analyzing, and interpreting complex biological datasets. The advances in AI, particularly in machine learning and deep learning, have been transforming the fields of genomics, planetary health, and agriculture. This article aims to unpack and explore the formidable range of possibilities and challenges that result from such transdisciplinary integration, and emphasizes its radically transformative potential for human and ecosystem health. The integration of these disciplines is also driving significant advancements in precision medicine and personalized health care. This presents an unprecedented opportunity to deepen our understanding of complex biological systems and advance the well-being of all life in planetary ecosystems. Notwithstanding in mind its sociotechnical, ethical, and critical policy challenges, the integration of genomics, multiomics, planetary health, and agriculture with AI-guided bioinformatics opens up vast opportunities for transnational collaborative efforts, data sharing, analysis, valorization, and interdisciplinary innovations in life sciences and integrative biology.
Collapse
Affiliation(s)
- Aakanksha Biswas
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| | - Aditi Kumari
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| | - D S Gaikwad
- Amity Institute of Organic Agriculture, Amity University, Noida, India
| | - Dhananjay K Pandey
- Amity Institute of Biotechnology, Amity University Jharkhand, Ranchi, India
| |
Collapse
|
26
|
Singh S, Sarma DK, Verma V, Nagpal R, Kumar M. Unveiling the future of metabolic medicine: omics technologies driving personalized solutions for precision treatment of metabolic disorders. Biochem Biophys Res Commun 2023; 682:1-20. [PMID: 37788525 DOI: 10.1016/j.bbrc.2023.09.064] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 10/05/2023]
Abstract
Metabolic disorders are increasingly prevalent worldwide, leading to high rates of morbidity and mortality. The variety of metabolic illnesses can be addressed through personalized medicine. The goal of personalized medicine is to give doctors the ability to anticipate the best course of treatment for patients with metabolic problems. By analyzing a patient's metabolomic, proteomic, genetic profile, and clinical data, physicians can identify relevant diagnostic, and predictive biomarkers and develop treatment plans and therapy for acute and chronic metabolic diseases. To achieve this goal, real-time modeling of clinical data and multiple omics is essential to pinpoint underlying biological mechanisms, risk factors, and possibly useful data to promote early diagnosis and prevention of complex diseases. Incorporating cutting-edge technologies like artificial intelligence and machine learning is crucial for consolidating diverse forms of data, examining multiple variables, establishing databases of clinical indicators to aid decision-making, and formulating ethical protocols to address concerns. This review article aims to explore the potential of personalized medicine utilizing omics approaches for the treatment of metabolic disorders. It focuses on the recent advancements in genomics, epigenomics, proteomics, metabolomics, and nutrigenomics, emphasizing their role in revolutionizing personalized medicine.
Collapse
Affiliation(s)
- Samradhi Singh
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal, 462030, Madhya Pradesh, India
| | - Devojit Kumar Sarma
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal, 462030, Madhya Pradesh, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, 226014, Uttar Pradesh, India
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, Tallahassee, FL, 32306, USA
| | - Manoj Kumar
- ICMR- National Institute for Research in Environmental Health, Bhopal Bypass Road, Bhouri, Bhopal, 462030, Madhya Pradesh, India.
| |
Collapse
|
27
|
Zhan C, Tang T, Wu E, Zhang Y, He M, Wu R, Bi C, Wang J, Zhang Y, Shen B. From multi-omics approaches to personalized medicine in myocardial infarction. Front Cardiovasc Med 2023; 10:1250340. [PMID: 37965091 PMCID: PMC10642346 DOI: 10.3389/fcvm.2023.1250340] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/17/2023] [Indexed: 11/16/2023] Open
Abstract
Myocardial infarction (MI) is a prevalent cardiovascular disease characterized by myocardial necrosis resulting from coronary artery ischemia and hypoxia, which can lead to severe complications such as arrhythmia, cardiac rupture, heart failure, and sudden death. Despite being a research hotspot, the etiological mechanism of MI remains unclear. The emergence and widespread use of omics technologies, including genomics, transcriptomics, proteomics, metabolomics, and other omics, have provided new opportunities for exploring the molecular mechanism of MI and identifying a large number of disease biomarkers. However, a single-omics approach has limitations in understanding the complex biological pathways of diseases. The multi-omics approach can reveal the interaction network among molecules at various levels and overcome the limitations of the single-omics approaches. This review focuses on the omics studies of MI, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, and other omics. The exploration extended into the domain of multi-omics integrative analysis, accompanied by a compilation of diverse online resources, databases, and tools conducive to these investigations. Additionally, we discussed the role and prospects of multi-omics approaches in personalized medicine, highlighting the potential for improving diagnosis, treatment, and prognosis of MI.
Collapse
Affiliation(s)
- Chaoying Zhan
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Tong Tang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Erman Wu
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yuxin Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- KeyLaboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Mengqiao He
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rongrong Wu
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Cheng Bi
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- KeyLaboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jiao Wang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yingbo Zhang
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Bairong Shen
- Department of Cardiology and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Gosline SJC, Veličković M, Pino JC, Day LZ, Attah IK, Swensen AC, Danna V, Posso C, Rodland KD, Chen J, Matthews CE, Campbell-Thompson M, Laskin J, Burnum-Johnson K, Zhu Y, Piehowski PD. Proteome Mapping of the Human Pancreatic Islet Microenvironment Reveals Endocrine-Exocrine Signaling Sphere of Influence. Mol Cell Proteomics 2023; 22:100592. [PMID: 37328065 PMCID: PMC10460696 DOI: 10.1016/j.mcpro.2023.100592] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 04/24/2023] [Accepted: 06/05/2023] [Indexed: 06/18/2023] Open
Abstract
The need for a clinically accessible method with the ability to match protein activity within heterogeneous tissues is currently unmet by existing technologies. Our proteomics sample preparation platform, named microPOTS (Microdroplet Processing in One pot for Trace Samples), can be used to measure relative protein abundance in micron-scale samples alongside the spatial location of each measurement, thereby tying biologically interesting proteins and pathways to distinct regions. However, given the smaller pixel/voxel number and amount of tissue measured, standard mass spectrometric analysis pipelines have proven inadequate. Here we describe how existing computational approaches can be adapted to focus on the specific biological questions asked in spatial proteomics experiments. We apply this approach to present an unbiased characterization of the human islet microenvironment comprising the entire complex array of cell types involved while maintaining spatial information and the degree of the islet's sphere of influence. We identify specific functional activity unique to the pancreatic islet cells and demonstrate how far their signature can be detected in the adjacent tissue. Our results show that we can distinguish pancreatic islet cells from the neighboring exocrine tissue environment, recapitulate known biological functions of islet cells, and identify a spatial gradient in the expression of RNA processing proteins within the islet microenvironment.
Collapse
Affiliation(s)
- Sara J C Gosline
- Pacific Northwest National Laboratories, Richland, Washington, USA
| | | | - James C Pino
- Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Le Z Day
- Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Isaac K Attah
- Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Adam C Swensen
- Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Vincent Danna
- Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Camilo Posso
- Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Karin D Rodland
- Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Jing Chen
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Clayton E Matthews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, Florida, USA
| | - Julia Laskin
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| | | | - Ying Zhu
- Pacific Northwest National Laboratories, Richland, Washington, USA
| | - Paul D Piehowski
- Pacific Northwest National Laboratories, Richland, Washington, USA.
| |
Collapse
|
29
|
Erdem C, Gross SM, Heiser LM, Birtwistle MR. MOBILE pipeline enables identification of context-specific networks and regulatory mechanisms. Nat Commun 2023; 14:3991. [PMID: 37414767 PMCID: PMC10326020 DOI: 10.1038/s41467-023-39729-2] [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: 07/27/2022] [Accepted: 06/27/2023] [Indexed: 07/08/2023] Open
Abstract
Robust identification of context-specific network features that control cellular phenotypes remains a challenge. We here introduce MOBILE (Multi-Omics Binary Integration via Lasso Ensembles) to nominate molecular features associated with cellular phenotypes and pathways. First, we use MOBILE to nominate mechanisms of interferon-γ (IFNγ) regulated PD-L1 expression. Our analyses suggest that IFNγ-controlled PD-L1 expression involves BST2, CLIC2, FAM83D, ACSL5, and HIST2H2AA3 genes, which were supported by prior literature. We also compare networks activated by related family members transforming growth factor-beta 1 (TGFβ1) and bone morphogenetic protein 2 (BMP2) and find that differences in ligand-induced changes in cell size and clustering properties are related to differences in laminin/collagen pathway activity. Finally, we demonstrate the broad applicability and adaptability of MOBILE by analyzing publicly available molecular datasets to investigate breast cancer subtype specific networks. Given the ever-growing availability of multi-omics datasets, we envision that MOBILE will be broadly useful for identification of context-specific molecular features and pathways.
Collapse
Affiliation(s)
- Cemal Erdem
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA
| | - Sean M Gross
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA.
| | - Marc R Birtwistle
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC, USA.
- Department of Bioengineering, Clemson University, Clemson, SC, USA.
| |
Collapse
|
30
|
Bruce JW, Park E, Magnano C, Horswill M, Richards A, Potts G, Hebert A, Islam N, Coon JJ, Gitter A, Sherer N, Ahlquist P. HIV-1 virological synapse formation enhances infection spread by dysregulating Aurora Kinase B. PLoS Pathog 2023; 19:e1011492. [PMID: 37459363 PMCID: PMC10374047 DOI: 10.1371/journal.ppat.1011492] [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: 10/26/2022] [Revised: 07/27/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
HIV-1 spreads efficiently through direct cell-to-cell transmission at virological synapses (VSs) formed by interactions between HIV-1 envelope proteins (Env) on the surface of infected cells and CD4 receptors on uninfected target cells. Env-CD4 interactions bring the infected and uninfected cellular membranes into close proximity and induce transport of viral and cellular factors to the VS for efficient virion assembly and HIV-1 transmission. Using novel, cell-specific stable isotope labeling and quantitative mass spectrometric proteomics, we identified extensive changes in the levels and phosphorylation states of proteins in HIV-1 infected producer cells upon mixing with CD4+ target cells under conditions inducing VS formation. These coculture-induced alterations involved multiple cellular pathways including transcription, TCR signaling and, unexpectedly, cell cycle regulation, and were dominated by Env-dependent responses. We confirmed the proteomic results using inhibitors targeting regulatory kinases and phosphatases in selected pathways identified by our proteomic analysis. Strikingly, inhibiting the key mitotic regulator Aurora kinase B (AURKB) in HIV-1 infected cells significantly increased HIV activity in cell-to-cell fusion and transmission but had little effect on cell-free infection. Consistent with this, we found that AURKB regulates the fusogenic activity of HIV-1 Env. In the Jurkat T cell line and primary T cells, HIV-1 Env:CD4 interaction also dramatically induced cell cycle-independent AURKB relocalization to the centromere, and this signaling required the long (150 aa) cytoplasmic C-terminal domain (CTD) of Env. These results imply that cytoplasmic/plasma membrane AURKB restricts HIV-1 envelope fusion, and that this restriction is overcome by Env CTD-induced AURKB relocalization. Taken together, our data reveal a new signaling pathway regulating HIV-1 cell-to-cell transmission and potential new avenues for therapeutic intervention through targeting the Env CTD and AURKB activity.
Collapse
Affiliation(s)
- James W. Bruce
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Eunju Park
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Chris Magnano
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Mark Horswill
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Alicia Richards
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Gregory Potts
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Alexander Hebert
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Nafisah Islam
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Joshua J. Coon
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Morgridge Institute for Research, Madison, Wisconsin, United States of America
| | - Anthony Gitter
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- Department of Computer Sciences, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Department of Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Nathan Sherer
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| | - Paul Ahlquist
- John and Jeanne Rowe Center for Research in Virology, Morgridge Institute for Research, Madison, Wisconsin, United States of America
- McArdle Laboratory for Cancer Research, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
- Institute for Molecular Virology, University of Wisconsin–Madison, Madison, Wisconsin, United States of America
| |
Collapse
|
31
|
Bachman JA, Gyori BM, Sorger PK. Automated assembly of molecular mechanisms at scale from text mining and curated databases. Mol Syst Biol 2023; 19:e11325. [PMID: 36938926 PMCID: PMC10167483 DOI: 10.15252/msb.202211325] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/21/2023] Open
Abstract
The analysis of omic data depends on machine-readable information about protein interactions, modifications, and activities as found in protein interaction networks, databases of post-translational modifications, and curated models of gene and protein function. These resources typically depend heavily on human curation. Natural language processing systems that read the primary literature have the potential to substantially extend knowledge resources while reducing the burden on human curators. However, machine-reading systems are limited by high error rates and commonly generate fragmentary and redundant information. Here, we describe an approach to precisely assemble molecular mechanisms at scale using multiple natural language processing systems and the Integrated Network and Dynamical Reasoning Assembler (INDRA). INDRA identifies full and partial overlaps in information extracted from published papers and pathway databases, uses predictive models to improve the reliability of machine reading, and thereby assembles individual pieces of information into non-redundant and broadly usable mechanistic knowledge. Using INDRA to create high-quality corpora of causal knowledge we show it is possible to extend protein-protein interaction databases and explain co-dependencies in the Cancer Dependency Map.
Collapse
Affiliation(s)
- John A Bachman
- Laboratory of Systems PharmacologyHarvard Medical SchoolBostonMAUSA
| | - Benjamin M Gyori
- Laboratory of Systems PharmacologyHarvard Medical SchoolBostonMAUSA
| | - Peter K Sorger
- Laboratory of Systems PharmacologyHarvard Medical SchoolBostonMAUSA
- Department of Systems BiologyHarvard Medical SchoolBostonMAUSA
| |
Collapse
|
32
|
Sahin AT, Yurtseven A, Dadmand S, Ozcan G, Akarlar BA, Kucuk NEO, Senturk A, Ergonul O, Can F, Tuncbag N, Ozlu N. Plasma proteomics identify potential severity biomarkers from COVID-19 associated network. Proteomics Clin Appl 2023; 17:e2200070. [PMID: 36217943 PMCID: PMC9874836 DOI: 10.1002/prca.202200070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/27/2022] [Accepted: 10/07/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE Coronavirus disease 2019 (COVID-19) continues to threaten public health globally. Severe acute respiratory coronavirus type 2 (SARS-CoV-2) infection-dependent alterations in the host cell signaling network may unveil potential target proteins and pathways for therapeutic strategies. In this study, we aim to define early severity biomarkers and monitor altered pathways in the course of SARS-CoV-2 infection. EXPERIMENTAL DESIGN We systematically analyzed plasma proteomes of COVID-19 patients from Turkey by using mass spectrometry. Different severity grades (moderate, severe, and critical) and periods of disease (early, inflammatory, and recovery) are monitored. Significant alterations in protein expressions are used to reconstruct the COVID-19 associated network that was further extended to connect viral and host proteins. RESULTS Across all COVID-19 patients, 111 differentially expressed proteins were found, of which 28 proteins were unique to our study mainly enriching in immunoglobulin production. By monitoring different severity grades and periods of disease, CLEC3B, MST1, and ITIH2 were identified as potential early predictors of COVID-19 severity. Most importantly, we extended the COVID-19 associated network with viral proteins and showed the connectedness of viral proteins with human proteins. The most connected viral protein ORF8, which has a role in immune evasion, targets many host proteins tightly connected to the deregulated human plasma proteins. CONCLUSIONS AND CLINICAL RELEVANCE Plasma proteomes from critical patients are intrinsically clustered in a distinct group than severe and moderate patients. Importantly, we did not recover any grouping based on the infection period, suggesting their distinct proteome even in the recovery phase. The new potential early severity markers can be further studied for their value in the clinics to monitor COVID-19 prognosis. Beyond the list of plasma proteins, our disease-associated network unravels altered pathways, and the possible therapeutic targets in SARS-CoV-2 infection by connecting human and viral proteins. Follow-up studies on the disease associated network that we propose here will be useful to determine molecular details of viral perturbation and to address how the infection affects human physiology.
Collapse
Affiliation(s)
- Ayse Tugce Sahin
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey.,Graduate School of Science and Engineering, Koc University, Istanbul, Turkey
| | - Ali Yurtseven
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey.,Graduate School of Science and Engineering, Koc University, Istanbul, Turkey
| | - Sina Dadmand
- Graduate School of Science and Engineering, Koc University, Istanbul, Turkey.,Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| | - Gulin Ozcan
- Koc University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey.,Graduate School of Health Sciences, Koc University, Istanbul, Turkey
| | - Busra A Akarlar
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey.,Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| | - Nazli Ezgi Ozkan Kucuk
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey.,Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey
| | - Aydanur Senturk
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey
| | - Onder Ergonul
- Graduate School of Health Sciences, Koc University, Istanbul, Turkey.,Koc University Is Bank Research Center for Infectious Diseases (KUISCID), Istanbul, Turkey
| | - Fusun Can
- Graduate School of Health Sciences, Koc University, Istanbul, Turkey.,Department of Infectious Diseases, School of Medicine, Koc University, Istanbul, Turkey
| | - Nurcan Tuncbag
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey.,Department of Medical Microbiology, School of Medicine, Koc University, Istanbul, Turkey.,Department of Medical Biology, School of Medicine, Koc University, Istanbul, Turkey
| | - Nurhan Ozlu
- Department of Molecular Biology and Genetics, Koc University, Istanbul, Turkey.,Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey.,Department of Medical Biology, School of Medicine, Koc University, Istanbul, Turkey
| |
Collapse
|
33
|
Paul I, Bolzan D, Youssef A, Gagnon KA, Hook H, Karemore G, Oliphant MUJ, Lin W, Liu Q, Phanse S, White C, Padhorny D, Kotelnikov S, Chen CS, Hu P, Denis GV, Kozakov D, Raught B, Siggers T, Wuchty S, Muthuswamy SK, Emili A. Parallelized multidimensional analytic framework applied to mammary epithelial cells uncovers regulatory principles in EMT. Nat Commun 2023; 14:688. [PMID: 36755019 PMCID: PMC9908882 DOI: 10.1038/s41467-023-36122-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023] Open
Abstract
A proper understanding of disease etiology will require longitudinal systems-scale reconstruction of the multitiered architecture of eukaryotic signaling. Here we combine state-of-the-art data acquisition platforms and bioinformatics tools to devise PAMAF, a workflow that simultaneously examines twelve omics modalities, i.e., protein abundance from whole-cells, nucleus, exosomes, secretome and membrane; N-glycosylation, phosphorylation; metabolites; mRNA, miRNA; and, in parallel, single-cell transcriptomes. We apply PAMAF in an established in vitro model of TGFβ-induced epithelial to mesenchymal transition (EMT) to quantify >61,000 molecules from 12 omics and 10 timepoints over 12 days. Bioinformatics analysis of this EMT-ExMap resource allowed us to identify; -topological coupling between omics, -four distinct cell states during EMT, -omics-specific kinetic paths, -stage-specific multi-omics characteristics, -distinct regulatory classes of genes, -ligand-receptor mediated intercellular crosstalk by integrating scRNAseq and subcellular proteomics, and -combinatorial drug targets (e.g., Hedgehog signaling and CAMK-II) to inhibit EMT, which we validate using a 3D mammary duct-on-a-chip platform. Overall, this study provides a resource on TGFβ signaling and EMT.
Collapse
Affiliation(s)
- Indranil Paul
- Department of Biochemistry, Boston University School of Medicine, Boston University, 71 East Concord Street, Boston, MA, 02118, USA
| | - Dante Bolzan
- Department of Computer Science, University of Miami, 1356 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Ahmed Youssef
- Graduate Program in Bioinformatics, Boston University, 24 Cummington Mall, Boston, MA, 02215, USA
| | - Keith A Gagnon
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA, 02215, USA
| | - Heather Hook
- Department of Biology, Boston University, 24 Cummington Mall, Boston, MA, 02115, USA
- Biological Design Center, Boston University, 610 Commonwealth Avenue, Boston, MA, 02215, USA
| | - Gopal Karemore
- Advanced Analytics, Novo Nordisk A/S, 2760, Måløv, Denmark
| | - Michael U J Oliphant
- Cancer Research Institute, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA
| | - Weiwei Lin
- Department of Biochemistry, Boston University School of Medicine, Boston University, 71 East Concord Street, Boston, MA, 02118, USA
| | - Qian Liu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, R3E 0J9, Canada
| | - Sadhna Phanse
- Department of Biochemistry, Boston University School of Medicine, Boston University, 71 East Concord Street, Boston, MA, 02118, USA
| | - Carl White
- Department of Biochemistry, Boston University School of Medicine, Boston University, 71 East Concord Street, Boston, MA, 02118, USA
| | - Dzmitry Padhorny
- Department of Applied Mathematics and Statistics, Stony Brook University, 11794, Stony Brook, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Sergei Kotelnikov
- Department of Applied Mathematics and Statistics, Stony Brook University, 11794, Stony Brook, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, 44 Cummington Mall, Boston, MA, 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, 3 Blackfan Circle, Boston, MA, 02115, USA
| | - Pingzhao Hu
- Department of Biochemistry, Western University, London, ON, N6A 5C1, Canada
| | - Gerald V Denis
- Boston Medical Center Cancer Center, Boston University, Boston University, 72 East Concord Street, Boston, MA, 02118, USA
| | - Dima Kozakov
- Department of Applied Mathematics and Statistics, Stony Brook University, 11794, Stony Brook, NY, USA
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Brian Raught
- Discovery Tower (TMDT), 101 College St, Rm. 9-701A, University of Toronto, Toronto, ON, M5G 1L7, Canada
| | - Trevor Siggers
- Department of Biology, Boston University, 24 Cummington Mall, Boston, MA, 02115, USA
- Biological Design Center, Boston University, 610 Commonwealth Avenue, Boston, MA, 02215, USA
| | - Stefan Wuchty
- Department of Computer Science, University of Miami, 1356 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Senthil K Muthuswamy
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Andrew Emili
- Department of Biochemistry, Boston University School of Medicine, Boston University, 71 East Concord Street, Boston, MA, 02118, USA.
- Department of Biology, Charles River Campus, Boston University, Life Science & Engineering (LSEB-602), 24 Cummington Mall, Boston, MA, 02215, USA.
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, USA.
| |
Collapse
|
34
|
Mapping the common gene networks that underlie related diseases. Nat Protoc 2023:10.1038/s41596-022-00797-1. [PMID: 36653526 DOI: 10.1038/s41596-022-00797-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 11/21/2022] [Indexed: 01/19/2023]
Abstract
A longstanding goal of biomedicine is to understand how alterations in molecular and cellular networks give rise to the spectrum of human diseases. For diseases with shared etiology, understanding the common causes allows for improved diagnosis of each disease, development of new therapies and more comprehensive identification of disease genes. Accordingly, this protocol describes how to evaluate the extent to which two diseases, each characterized by a set of mapped genes, are colocalized in a reference gene interaction network. This procedure uses network propagation to measure the network 'distance' between gene sets. For colocalized diseases, the network can be further analyzed to extract common gene communities at progressive granularities. In particular, we show how to: (1) obtain input gene sets and a reference gene interaction network; (2) identify common subnetworks of genes that encompass or are in close proximity to all gene sets; (3) use multiscale community detection to identify systems and pathways represented by each common subnetwork to generate a network colocalized systems map; (4) validate identified genes and systems using a mouse variant database; and (5) visualize and further investigate select genes, interactions and systems for relevance to phenotype(s) of interest. We demonstrate the utility of this approach by identifying shared biological mechanisms underlying autism and congenital heart disease. However, this protocol is general and can be applied to any gene sets attributed to diseases or other phenotypes with suspected joint association. A typical NetColoc run takes less than an hour. Software and documentation are available at https://github.com/ucsd-ccbb/NetColoc .
Collapse
|
35
|
Magnano CS, Gitter A. Graph algorithms for predicting subcellular localization at the pathway level. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2023; 28:145-156. [PMID: 36540972 PMCID: PMC9817068 DOI: 10.1142/9789811270611_0014] [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] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Protein subcellular localization is an important factor in normal cellular processes and disease. While many protein localization resources treat it as static, protein localization is dynamic and heavily influenced by biological context. Biological pathways are graphs that represent a specific biological context and can be inferred from large-scale data. We develop graph algorithms to predict the localization of all interactions in a biological pathway as an edge-labeling task. We compare a variety of models including graph neural networks, probabilistic graphical models, and discriminative classifiers for predicting localization annotations from curated pathway databases. We also perform a case study where we construct biological pathways and predict localizations of human fibroblasts undergoing viral infection. Pathway localization prediction is a promising approach for integrating publicly available localization data into the analysis of large-scale biological data.
Collapse
Affiliation(s)
- Chris S Magnano
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
- Center for Computational Biomedicine, Harvard Medical School, Boston, MA, USA
| | - Anthony Gitter
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
36
|
Galindez G, Sadegh S, Baumbach J, Kacprowski T, List M. Network-based approaches for modeling disease regulation and progression. Comput Struct Biotechnol J 2022; 21:780-795. [PMID: 36698974 PMCID: PMC9841310 DOI: 10.1016/j.csbj.2022.12.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 12/14/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Molecular interaction networks lay the foundation for studying how biological functions are controlled by the complex interplay of genes and proteins. Investigating perturbed processes using biological networks has been instrumental in uncovering mechanisms that underlie complex disease phenotypes. Rapid advances in omics technologies have prompted the generation of high-throughput datasets, enabling large-scale, network-based analyses. Consequently, various modeling techniques, including network enrichment, differential network extraction, and network inference, have proven to be useful for gaining new mechanistic insights. We provide an overview of recent network-based methods and their core ideas to facilitate the discovery of disease modules or candidate mechanisms. Knowledge generated from these computational efforts will benefit biomedical research, especially drug development and precision medicine. We further discuss current challenges and provide perspectives in the field, highlighting the need for more integrative and dynamic network approaches to model disease development and progression.
Collapse
Affiliation(s)
- Gihanna Galindez
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of Technische Universität Braunschweig and Hannover Medical School, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), TU Braunschweig, Braunschweig, Germany
| | - Sepideh Sadegh
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
| | - Jan Baumbach
- Institute for Computational Systems Biology, University of Hamburg, Hamburg, Germany
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Tim Kacprowski
- Division Data Science in Biomedicine, Peter L. Reichertz Institute for Medical Informatics of Technische Universität Braunschweig and Hannover Medical School, Braunschweig, Germany
- Braunschweig Integrated Centre of Systems Biology (BRICS), TU Braunschweig, Braunschweig, Germany
| | - Markus List
- Chair of Experimental Bioinformatics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| |
Collapse
|
37
|
Athieniti E, Spyrou GM. A guide to multi-omics data collection and integration for translational medicine. Comput Struct Biotechnol J 2022; 21:134-149. [PMID: 36544480 PMCID: PMC9747357 DOI: 10.1016/j.csbj.2022.11.050] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/02/2022] Open
Abstract
The emerging high-throughput technologies have led to the shift in the design of translational medicine projects towards collecting multi-omics patient samples and, consequently, their integrated analysis. However, the complexity of integrating these datasets has triggered new questions regarding the appropriateness of the available computational methods. Currently, there is no clear consensus on the best combination of omics to include and the data integration methodologies required for their analysis. This article aims to guide the design of multi-omics studies in the field of translational medicine regarding the types of omics and the integration method to choose. We review articles that perform the integration of multiple omics measurements from patient samples. We identify five objectives in translational medicine applications: (i) detect disease-associated molecular patterns, (ii) subtype identification, (iii) diagnosis/prognosis, (iv) drug response prediction, and (v) understand regulatory processes. We describe common trends in the selection of omic types combined for different objectives and diseases. To guide the choice of data integration tools, we group them into the scientific objectives they aim to address. We describe the main computational methods adopted to achieve these objectives and present examples of tools. We compare tools based on how they deal with the computational challenges of data integration and comment on how they perform against predefined objective-specific evaluation criteria. Finally, we discuss examples of tools for downstream analysis and further extraction of novel insights from multi-omics datasets.
Collapse
Affiliation(s)
- Efi Athieniti
- Department of Bioinformatics, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371 Ayios Dometios, Nicosia, Cyprus
| | - George M. Spyrou
- Department of Bioinformatics, The Cyprus Institute of Neurology and Genetics, 6 Iroon Avenue, 2371 Ayios Dometios, Nicosia, Cyprus
| |
Collapse
|
38
|
Singh A, Ambaru B, Bandsode V, Ahmed N. Panomics to decode virulence and fitness in Gram-negative bacteria. Front Cell Infect Microbiol 2022; 12:1061596. [PMID: 36478674 PMCID: PMC9719987 DOI: 10.3389/fcimb.2022.1061596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 10/26/2022] [Indexed: 11/22/2022] Open
|
39
|
Agamah FE, Bayjanov JR, Niehues A, Njoku KF, Skelton M, Mazandu GK, Ederveen THA, Mulder N, Chimusa ER, 't Hoen PAC. Computational approaches for network-based integrative multi-omics analysis. Front Mol Biosci 2022; 9:967205. [PMID: 36452456 PMCID: PMC9703081 DOI: 10.3389/fmolb.2022.967205] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 10/20/2022] [Indexed: 08/27/2023] Open
Abstract
Advances in omics technologies allow for holistic studies into biological systems. These studies rely on integrative data analysis techniques to obtain a comprehensive view of the dynamics of cellular processes, and molecular mechanisms. Network-based integrative approaches have revolutionized multi-omics analysis by providing the framework to represent interactions between multiple different omics-layers in a graph, which may faithfully reflect the molecular wiring in a cell. Here we review network-based multi-omics/multi-modal integrative analytical approaches. We classify these approaches according to the type of omics data supported, the methods and/or algorithms implemented, their node and/or edge weighting components, and their ability to identify key nodes and subnetworks. We show how these approaches can be used to identify biomarkers, disease subtypes, crosstalk, causality, and molecular drivers of physiological and pathological mechanisms. We provide insight into the most appropriate methods and tools for research questions as showcased around the aetiology and treatment of COVID-19 that can be informed by multi-omics data integration. We conclude with an overview of challenges associated with multi-omics network-based analysis, such as reproducibility, heterogeneity, (biological) interpretability of the results, and we highlight some future directions for network-based integration.
Collapse
Affiliation(s)
- Francis E. Agamah
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Jumamurat R. Bayjanov
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anna Niehues
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Kelechi F. Njoku
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Michelle Skelton
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Gaston K. Mazandu
- Division of Human Genetics, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- African Institute for Mathematical Sciences, Cape Town, South Africa
| | - Thomas H. A. Ederveen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nicola Mulder
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, CIDRI-Africa Wellcome Trust Centre, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle, United Kingdom
| | - Peter A. C. 't Hoen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
40
|
Kutnu M, İşlerel ET, Tunçbağ N, Özcengiz G. Comparative biological network analysis for differentially expressed proteins as a function of bacilysin biosynthesis in Bacillus subtilis. Integr Biol (Camb) 2022; 14:99-110. [PMID: 35901454 DOI: 10.1093/intbio/zyac010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 12/07/2021] [Accepted: 01/05/2022] [Indexed: 06/15/2023]
Abstract
The Gram-positive bacterium Bacillus subtilis produces a diverse range of secondary metabolites with different structures and activities. Among them, bacilysin is an enzymatically synthesized dipeptide that consists of L-alanine and L-anticapsin. Previous research by our group has suggested bacilysin's role as a pleiotropic molecule in its producer, B. subtilis PY79. However, the nature of protein interactions in the absence of bacilysin has not been defined. In the present work, we constructed a protein-protein interaction subnetwork by using Omics Integrator based on our recent comparative proteomics data obtained from a bacilysin-silenced strain, OGU1. Functional enrichment analyses on the resulting networks pointed to certain putatively perturbed pathways such as citrate cycle, quorum sensing and secondary metabolite biosynthesis. Various molecules, which were absent from the experimental data, were included in the final network. We believe that this study can guide further experiments in the identification and confirmation of protein-protein interactions in B. subtilis.
Collapse
Affiliation(s)
- Meltem Kutnu
- Department of Biological Sciences/Molecular Biology and Genetics, Middle East Technical University, Ankara 06800, Turkey
| | - Elif Tekin İşlerel
- Department of Biological Sciences/Molecular Biology and Genetics, Middle East Technical University, Ankara 06800, Turkey
- Department of Medical Microbiology, Faculty of Medicine, Maltepe University, Istanbul 34857, Turkey
| | - Nurcan Tunçbağ
- Department of Chemical and Biological Engineering, Koc University, Istanbul 34450, Turkey
| | - Gülay Özcengiz
- Department of Biological Sciences/Molecular Biology and Genetics, Middle East Technical University, Ankara 06800, Turkey
| |
Collapse
|
41
|
Gosline SJC, Tognon C, Nestor M, Joshi S, Modak R, Damnernsawad A, Posso C, Moon J, Hansen JR, Hutchinson-Bunch C, Pino JC, Gritsenko MA, Weitz KK, Traer E, Tyner J, Druker B, Agarwal A, Piehowski P, McDermott JE, Rodland K. Proteomic and phosphoproteomic measurements enhance ability to predict ex vivo drug response in AML. Clin Proteomics 2022; 19:30. [PMID: 35896960 PMCID: PMC9327422 DOI: 10.1186/s12014-022-09367-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 06/22/2022] [Indexed: 11/23/2022] Open
Abstract
Acute Myeloid Leukemia (AML) affects 20,000 patients in the US annually with a five-year survival rate of approximately 25%. One reason for the low survival rate is the high prevalence of clonal evolution that gives rise to heterogeneous sub-populations of leukemic cells with diverse mutation spectra, which eventually leads to disease relapse. This genetic heterogeneity drives the activation of complex signaling pathways that is reflected at the protein level. This diversity makes it difficult to treat AML with targeted therapy, requiring custom patient treatment protocols tailored to each individual's leukemia. Toward this end, the Beat AML research program prospectively collected genomic and transcriptomic data from over 1000 AML patients and carried out ex vivo drug sensitivity assays to identify genomic signatures that could predict patient-specific drug responses. However, there are inherent weaknesses in using only genetic and transcriptomic measurements as surrogates of drug response, particularly the absence of direct information about phosphorylation-mediated signal transduction. As a member of the Clinical Proteomic Tumor Analysis Consortium, we have extended the molecular characterization of this cohort by collecting proteomic and phosphoproteomic measurements from a subset of these patient samples (38 in total) to evaluate the hypothesis that proteomic signatures can improve the ability to predict response to 26 drugs in AML ex vivo samples. In this work we describe our systematic, multi-omic approach to evaluate proteomic signatures of drug response and compare protein levels to other markers of drug response such as mutational patterns. We explore the nuances of this approach using two drugs that target key pathways activated in AML: quizartinib (FLT3) and trametinib (Ras/MEK), and show how patient-derived signatures can be interpreted biologically and validated in cell lines. In conclusion, this pilot study demonstrates strong promise for proteomics-based patient stratification to assess drug sensitivity in AML.
Collapse
Affiliation(s)
| | - Cristina Tognon
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | | | - Sunil Joshi
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Rucha Modak
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alisa Damnernsawad
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Camilo Posso
- Pacific Northwest National Laboratory, Seattle, WA, USA
| | - Jamie Moon
- Pacific Northwest National Laboratory, Seattle, WA, USA
| | | | | | - James C Pino
- Pacific Northwest National Laboratory, Seattle, WA, USA
| | | | - Karl K Weitz
- Pacific Northwest National Laboratory, Seattle, WA, USA
| | - Elie Traer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Brian Druker
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
- Division of Oncological Sciences, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | | | - Jason E McDermott
- Pacific Northwest National Laboratory, Seattle, WA, USA
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, USA
| | - Karin Rodland
- Pacific Northwest National Laboratory, Seattle, WA, USA.
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
42
|
Hammelman J, Patel T, Closser M, Wichterle H, Gifford D. Ranking reprogramming factors for cell differentiation. Nat Methods 2022; 19:812-822. [PMID: 35710610 PMCID: PMC10460539 DOI: 10.1038/s41592-022-01522-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 05/13/2022] [Indexed: 12/16/2022]
Abstract
Transcription factor over-expression is a proven method for reprogramming cells to a desired cell type for regenerative medicine and therapeutic discovery. However, a general method for the identification of reprogramming factors to create an arbitrary cell type is an open problem. Here we examine the success rate of methods and data for differentiation by testing the ability of nine computational methods (CellNet, GarNet, EBseq, AME, DREME, HOMER, KMAC, diffTF and DeepAccess) to discover and rank candidate factors for eight target cell types with known reprogramming solutions. We compare methods that use gene expression, biological networks and chromatin accessibility data, and comprehensively test parameter and preprocessing of input data to optimize performance. We find the best factor identification methods can identify an average of 50-60% of reprogramming factors within the top ten candidates, and methods that use chromatin accessibility perform the best. Among the chromatin accessibility methods, complex methods DeepAccess and diffTF have higher correlation with the ranked significance of transcription factor candidates within reprogramming protocols for differentiation. We provide evidence that AME and diffTF are optimal methods for transcription factor recovery that will allow for systematic prioritization of transcription factor candidates to aid in the design of new reprogramming protocols.
Collapse
Affiliation(s)
- Jennifer Hammelman
- Computational and Systems Biology, MIT, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, USA
| | - Tulsi Patel
- Departments of Pathology and Cell Biology, Neuroscience, Rehabilitation and Regenerative Medicine (in Neurology), Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael Closser
- Departments of Pathology and Cell Biology, Neuroscience, Rehabilitation and Regenerative Medicine (in Neurology), Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY, USA
| | - Hynek Wichterle
- Departments of Pathology and Cell Biology, Neuroscience, Rehabilitation and Regenerative Medicine (in Neurology), Columbia University Irving Medical Center, New York, NY, USA
- Center for Motor Neuron Biology and Disease, Columbia University Irving Medical Center, New York, NY, USA
- Columbia Stem Cell Initiative, Columbia University Irving Medical Center, New York, NY, USA
| | - David Gifford
- Computational and Systems Biology, MIT, Cambridge, MA, USA.
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA, USA.
- Department of Biological Engineering, MIT, Cambridge, MA, USA.
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA.
| |
Collapse
|
43
|
Gul H, Selvi S, Yilmaz F, Ozcelik G, Olfaz‐Aslan S, Yazan S, Tiryaki B, Gul S, Yurtseven A, Kavakli IH, Ozlu N, Ozturk N. Proteome analysis of the circadian clock protein PERIOD2. Proteins 2022; 90:1315-1330. [DOI: 10.1002/prot.26314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/25/2022] [Accepted: 01/29/2022] [Indexed: 12/17/2022]
Affiliation(s)
- Huseyin Gul
- Department of Molecular Biology and Genetics Gebze Technical University Gebze Kocaeli Turkey
| | - Saba Selvi
- Department of Molecular Biology and Genetics Gebze Technical University Gebze Kocaeli Turkey
| | - Fatma Yilmaz
- Department of Molecular Biology and Genetics Gebze Technical University Gebze Kocaeli Turkey
| | - Gozde Ozcelik
- Department of Molecular Biology and Genetics Gebze Technical University Gebze Kocaeli Turkey
| | - Senanur Olfaz‐Aslan
- Department of Molecular Biology and Genetics Gebze Technical University Gebze Kocaeli Turkey
| | - Seyma Yazan
- Department of Molecular Biology and Genetics Gebze Technical University Gebze Kocaeli Turkey
| | - Busra Tiryaki
- Department of Molecular Biology and Genetics Gebze Technical University Gebze Kocaeli Turkey
| | - Seref Gul
- Department of Biology Istanbul University Istanbul Turkey
| | - Ali Yurtseven
- Department of Molecular Biology and Genetics Koc University Istanbul Turkey
| | - Ibrahim Halil Kavakli
- Department of Molecular Biology and Genetics Koc University Istanbul Turkey
- Department of Chemical and Biological Engineering Koc University Istanbul Turkey
| | - Nurhan Ozlu
- Department of Molecular Biology and Genetics Koc University Istanbul Turkey
| | - Nuri Ozturk
- Department of Molecular Biology and Genetics Gebze Technical University Gebze Kocaeli Turkey
| |
Collapse
|
44
|
Functional stratification of cancer drugs through integrated network similarity. NPJ Syst Biol Appl 2022; 8:11. [PMID: 35440787 PMCID: PMC9018743 DOI: 10.1038/s41540-022-00219-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/21/2022] [Indexed: 11/30/2022] Open
Abstract
Drugs not only perturb their immediate protein targets but also modulate multiple signaling pathways. In this study, we explored networks modulated by several drugs across multiple cancer cell lines by integrating their targets with transcriptomic and phosphoproteomic data. As a result, we obtained 236 reconstructed networks covering five cell lines and 70 drugs. A rigorous topological and pathway analysis showed that chemically and functionally different drugs may modulate overlapping networks. Additionally, we revealed a set of tumor-specific hidden pathways with the help of drug network models that are not detectable from the initial data. The difference in the target selectivity of the drugs leads to disjoint networks despite sharing a similar mechanism of action, e.g., HDAC inhibitors. We also used the reconstructed network models to study potential drug combinations based on the topological separation and found literature evidence for a set of drug pairs. Overall, network-level exploration of drug-modulated pathways and their deep comparison may potentially help optimize treatment strategies and suggest new drug combinations.
Collapse
|
45
|
Winkler S, Winkler I, Figaschewski M, Tiede T, Nordheim A, Kohlbacher O. De novo identification of maximally deregulated subnetworks based on multi-omics data with DeRegNet. BMC Bioinformatics 2022; 23:139. [PMID: 35439941 PMCID: PMC9020058 DOI: 10.1186/s12859-022-04670-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 03/29/2022] [Indexed: 12/14/2022] Open
Abstract
Background With a growing amount of (multi-)omics data being available, the extraction of knowledge from these datasets is still a difficult problem. Classical enrichment-style analyses require predefined pathways or gene sets that are tested for significant deregulation to assess whether the pathway is functionally involved in the biological process under study. De novo identification of these pathways can reduce the bias inherent in predefined pathways or gene sets. At the same time, the definition and efficient identification of these pathways de novo from large biological networks is a challenging problem. Results We present a novel algorithm, DeRegNet, for the identification of maximally deregulated subnetworks on directed graphs based on deregulation scores derived from (multi-)omics data. DeRegNet can be interpreted as maximum likelihood estimation given a certain probabilistic model for de-novo subgraph identification. We use fractional integer programming to solve the resulting combinatorial optimization problem. We can show that the approach outperforms related algorithms on simulated data with known ground truths. On a publicly available liver cancer dataset we can show that DeRegNet can identify biologically meaningful subgraphs suitable for patient stratification. DeRegNet can also be used to find explicitly multi-omics subgraphs which we demonstrate by presenting subgraphs with consistent methylation-transcription patterns. DeRegNet is freely available as open-source software. Conclusion The proposed algorithmic framework and its available implementation can serve as a valuable heuristic hypothesis generation tool contextualizing omics data within biomolecular networks.
Collapse
Affiliation(s)
- Sebastian Winkler
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany. .,International Max Planck Research School (IMPRS) "From Molecules to Organism", Tübingen, Germany.
| | - Ivana Winkler
- International Max Planck Research School (IMPRS) "From Molecules to Organism", Tübingen, Germany.,Interfaculty Institute for Cell Biology (IFIZ), University of Tuebingen, Tübingen, Germany.,German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mirjam Figaschewski
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany
| | - Thorsten Tiede
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany
| | - Alfred Nordheim
- Interfaculty Institute for Cell Biology (IFIZ), University of Tuebingen, Tübingen, Germany.,Leibniz Institute on Aging (FLI), Jena, Germany
| | - Oliver Kohlbacher
- Applied Bioinformatics, Department of Computer Science, University of Tuebingen, Tübingen, Germany.,Institute for Bioinformatics and Medical Informatics, University of Tuebingen, Tübingen, Germany.,Translational Bioinformatics, University Hospital Tuebingen, Tübingen, Germany
| |
Collapse
|
46
|
Kaiser T, Jahansouz C, Staley C. Network-based approaches for the investigation of microbial community structure and function using metagenomics-based data. Future Microbiol 2022; 17:621-631. [PMID: 35360922 DOI: 10.2217/fmb-2021-0219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Network-based approaches offer a powerful framework to evaluate microbial community organization and function as it relates to a variety of environmental processes. Emerging studies are exploring network theory as a method for data integration that is likely to be critical for the integration of 'omics' data using systems biology approaches. Intricacies of network theory and methodological and computational complexities in network construction, however, impede the use of these tools for translational science. We provide a perspective on the methods of network construction, interpretation and emerging uses for these techniques in understanding host-microbiota interactions.
Collapse
Affiliation(s)
- Thomas Kaiser
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA.,Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
| | - Cyrus Jahansouz
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher Staley
- Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA.,Biotechnology Institute, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
47
|
Paulsen B, Velasco S, Kedaigle AJ, Pigoni M, Quadrato G, Deo AJ, Adiconis X, Uzquiano A, Sartore R, Yang SM, Simmons SK, Symvoulidis P, Kim K, Tsafou K, Podury A, Abbate C, Tucewicz A, Smith SN, Albanese A, Barrett L, Sanjana NE, Shi X, Chung K, Lage K, Boyden ES, Regev A, Levin JZ, Arlotta P. Autism genes converge on asynchronous development of shared neuron classes. Nature 2022; 602:268-273. [PMID: 35110736 PMCID: PMC8852827 DOI: 10.1038/s41586-021-04358-6] [Citation(s) in RCA: 223] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 12/14/2021] [Indexed: 02/08/2023]
Abstract
Genetic risk for autism spectrum disorder (ASD) is associated with hundreds of genes spanning a wide range of biological functions1-6. The alterations in the human brain resulting from mutations in these genes remain unclear. Furthermore, their phenotypic manifestation varies across individuals7,8. Here we used organoid models of the human cerebral cortex to identify cell-type-specific developmental abnormalities that result from haploinsufficiency in three ASD risk genes-SUV420H1 (also known as KMT5B), ARID1B and CHD8-in multiple cell lines from different donors, using single-cell RNA-sequencing (scRNA-seq) analysis of more than 745,000 cells and proteomic analysis of individual organoids, to identify phenotypic convergence. Each of the three mutations confers asynchronous development of two main cortical neuronal lineages-γ-aminobutyric-acid-releasing (GABAergic) neurons and deep-layer excitatory projection neurons-but acts through largely distinct molecular pathways. Although these phenotypes are consistent across cell lines, their expressivity is influenced by the individual genomic context, in a manner that is dependent on both the risk gene and the developmental defect. Calcium imaging in intact organoids shows that these early-stage developmental changes are followed by abnormal circuit activity. This research uncovers cell-type-specific neurodevelopmental abnormalities that are shared across ASD risk genes and are finely modulated by human genomic context, finding convergence in the neurobiological basis of how different risk genes contribute to ASD pathology.
Collapse
Affiliation(s)
- Bruna Paulsen
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Silvia Velasco
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, Victoria, Australia.
| | - Amanda J Kedaigle
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Martina Pigoni
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Giorgia Quadrato
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Anthony J Deo
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Psychiatry, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, USA
- Rutgers University Behavioral Health Care, Piscataway, NJ, USA
| | - Xian Adiconis
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ana Uzquiano
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rafaela Sartore
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sung Min Yang
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sean K Simmons
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Panagiotis Symvoulidis
- MIT Center for Neurobiological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Kwanho Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kalliopi Tsafou
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Archana Podury
- MIT Center for Neurobiological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Harvard-MIT Health Sciences & Technology Program (HST), Harvard Medical School, Boston, MA, USA
| | - Catherine Abbate
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashley Tucewicz
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samantha N Smith
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alexandre Albanese
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Lindy Barrett
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Neville E Sanjana
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- New York Genome Center, New York, NY, USA
- Department of Biology, New York University, New York, NY, USA
| | - Xi Shi
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kwanghun Chung
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Picower Institute for Learning and Memory, Departments of Chemical Engineering and Brain and Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- New York Genome Center, New York, NY, USA
| | - Kasper Lage
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Surgery and Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Edward S Boyden
- MIT Center for Neurobiological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Harvard-MIT Health Sciences & Technology Program (HST), Harvard Medical School, Boston, MA, USA
- McGovern Institute for Brain Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Howard Hughes Medical Institute, MIT, Cambridge, MA, USA
- Department of Brain of Cognitive Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Media Arts and Sciences, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
- Genentech, South San Francisco, CA, USA
| | - Joshua Z Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
48
|
Blum BC, Lin W, Lawton ML, Liu Q, Kwan J, Turcinovic I, Hekman R, Hu P, Emili A. Multiomic Metabolic Enrichment Network Analysis Reveals Metabolite-Protein Physical Interaction Subnetworks Altered in Cancer. Mol Cell Proteomics 2022; 21:100189. [PMID: 34933084 PMCID: PMC8761777 DOI: 10.1016/j.mcpro.2021.100189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/04/2021] [Accepted: 12/16/2021] [Indexed: 11/25/2022] Open
Abstract
Metabolism is recognized as an important driver of cancer progression and other complex diseases, but global metabolite profiling remains a challenge. Protein expression profiling is often a poor proxy since existing pathway enrichment models provide an incomplete mapping between the proteome and metabolism. To overcome these gaps, we introduce multiomic metabolic enrichment network analysis (MOMENTA), an integrative multiomic data analysis framework for more accurately deducing metabolic pathway changes from proteomics data alone in a gene set analysis context by leveraging protein interaction networks to extend annotated metabolic models. We apply MOMENTA to proteomic data from diverse cancer cell lines and human tumors to demonstrate its utility at revealing variation in metabolic pathway activity across cancer types, which we verify using independent metabolomics measurements. The novel metabolic networks we uncover in breast cancer and other tumors are linked to clinical outcomes, underscoring the pathophysiological relevance of the findings.
Collapse
Affiliation(s)
- Benjamin C Blum
- Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA; Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Weiwei Lin
- Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA; Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Matthew L Lawton
- Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA; Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Qian Liu
- Departments of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Julian Kwan
- Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA; Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Isabella Turcinovic
- Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA
| | - Ryan Hekman
- Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA; Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Pingzhao Hu
- Departments of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew Emili
- Center for Network Systems Biology, Boston University, Boston, Massachusetts, USA; Department of Biochemistry, Boston University School of Medicine, Boston, Massachusetts, USA; Department of Biology, Boston University, Boston, Massachusetts, USA.
| |
Collapse
|
49
|
Li J, Lim RG, Kaye JA, Dardov V, Coyne AN, Wu J, Milani P, Cheng A, Thompson TG, Ornelas L, Frank A, Adam M, Banuelos MG, Casale M, Cox V, Escalante-Chong R, Daigle JG, Gomez E, Hayes L, Holewenski R, Lei S, Lenail A, Lima L, Mandefro B, Matlock A, Panther L, Patel-Murray NL, Pham J, Ramamoorthy D, Sachs K, Shelley B, Stocksdale J, Trost H, Wilhelm M, Venkatraman V, Wassie BT, Wyman S, Yang S, Van Eyk JE, Lloyd TE, Finkbeiner S, Fraenkel E, Rothstein JD, Sareen D, Svendsen CN, Thompson LM. An integrated multi-omic analysis of iPSC-derived motor neurons from C9ORF72 ALS patients. iScience 2021; 24:103221. [PMID: 34746695 PMCID: PMC8554488 DOI: 10.1016/j.isci.2021.103221] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/29/2021] [Accepted: 09/30/2021] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases are challenging for systems biology because of the lack of reliable animal models or patient samples at early disease stages. Induced pluripotent stem cells (iPSCs) could address these challenges. We investigated DNA, RNA, epigenetics, and proteins in iPSC-derived motor neurons from patients with ALS carrying hexanucleotide expansions in C9ORF72. Using integrative computational methods combining all omics datasets, we identified novel and known dysregulated pathways. We used a C9ORF72 Drosophila model to distinguish pathways contributing to disease phenotypes from compensatory ones and confirmed alterations in some pathways in postmortem spinal cord tissue of patients with ALS. A different differentiation protocol was used to derive a separate set of C9ORF72 and control motor neurons. Many individual -omics differed by protocol, but some core dysregulated pathways were consistent. This strategy of analyzing patient-specific neurons provides disease-related outcomes with small numbers of heterogeneous lines and reduces variation from single-omics to elucidate network-based signatures.
Collapse
Affiliation(s)
- Jonathan Li
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ryan G. Lim
- UCI MIND, University of California, Irvine, CA 92697, USA
| | - Julia A. Kaye
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Victoria Dardov
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alyssa N. Coyne
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Jie Wu
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA
| | - Pamela Milani
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Andrew Cheng
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | | | - Loren Ornelas
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Aaron Frank
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Miriam Adam
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Maria G. Banuelos
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Malcolm Casale
- UCI MIND, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Veerle Cox
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Renan Escalante-Chong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - J. Gavin Daigle
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Emilda Gomez
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Lindsey Hayes
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Ronald Holewenski
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Susan Lei
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Alex Lenail
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Leandro Lima
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Berhan Mandefro
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Andrea Matlock
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Lindsay Panther
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | | | - Jacqueline Pham
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Divya Ramamoorthy
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Karen Sachs
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Brandon Shelley
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Jennifer Stocksdale
- UCI MIND, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
| | - Hannah Trost
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Mark Wilhelm
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Vidya Venkatraman
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Brook T. Wassie
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Stacia Wyman
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697, USA
| | - Stephanie Yang
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | | | - Jennifer E. Van Eyk
- Advanced Clinical Biosystems Research Institute, The Barbra Streisand Heart Center, The Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Thomas E. Lloyd
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics and the Taube/Koret Center for Neurodegenerative Disease, Gladstone Institutes, University of California, San Francisco, San Francisco, CA 94158, USA
- Departments of Neurology and Physiology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ernest Fraenkel
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jeffrey D. Rothstein
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
- Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MA 212056, USA
| | - Dhruv Sareen
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Clive N. Svendsen
- The Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| | - Leslie M. Thompson
- UCI MIND, University of California, Irvine, CA 92697, USA
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, USA
- Sue and Bill Gross Stem Cell Center, University of California, Irvine, CA 92697, USA
| |
Collapse
|
50
|
Qiu S, Lavallée-Adam M, Côté M. Proximity Interactome Map of the Vac14-Fig4 Complex Using BioID. J Proteome Res 2021; 20:4959-4973. [PMID: 34554760 DOI: 10.1021/acs.jproteome.1c00408] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Conversion between phosphatidylinositol-3-phosphate and phosphatidylinositol-3,5-bisphosphate on endosomal membranes is critical for the maturation of early endosomes to late endosomes/lysosomes and is regulated by the PIKfyve-Vac14-Fig4 complex. Despite the importance of this complex for endosomal homeostasis and vesicular trafficking, there is little known about how its activity is regulated or how it interacts with other cellular proteins. Here, we screened for the cellular interactome of Vac14 and Fig4 using proximity-dependent biotin labeling (BioID). After independently screening the interactomes of Vac14 and Fig4, we identified 89 high-confidence protein hits shared by both proteins. Network analysis of these hits revealed pathways with known involvement of the PIKfyve-Vac14-Fig4 complex, including vesicular organization and PI3K/Akt signaling, as well as novel pathways including cell cycle and mitochondrial regulation. We also identified subunits of coatomer complex I (COPI), a Golgi-associated complex with an emerging role in endosomal dynamics. Using proximity ligation assays, we validated the interaction between Vac14 and COPI subunit COPB1 and between Vac14 and Arf1, a GTPase required for COPI assembly. In summary, this study used BioID to comprehensively map the Vac14-Fig4 interactome, revealing potential roles for these proteins in diverse cellular processes and pathways, including preliminary evidence of an interaction between Vac14 and COPI. Data are available via ProteomeXchange with the identifier PXD027917.
Collapse
Affiliation(s)
- Shirley Qiu
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada.,Ottawa Institute of Systems Biology, University of Ottawa, Ottawa K1H 8M5, Canada.,Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Mathieu Lavallée-Adam
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada.,Ottawa Institute of Systems Biology, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa K1H 8M5, Canada.,Ottawa Institute of Systems Biology, University of Ottawa, Ottawa K1H 8M5, Canada.,Centre for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa K1H 8M5, Canada
| |
Collapse
|