1
|
Awasthi BW, Paulo JA, Burkhart DL, Smith IR, Collins RL, Harper JW, Gygi SP, Haigis KM. The network response to Egf is tissue-specific. iScience 2025; 28:112146. [PMID: 40171493 PMCID: PMC11960661 DOI: 10.1016/j.isci.2025.112146] [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: 04/22/2024] [Revised: 07/29/2024] [Accepted: 02/27/2025] [Indexed: 04/03/2025] Open
Abstract
Epidermal growth factor receptor (Egfr)-driven signaling regulates fundamental homeostatic processes. Dysregulated signaling via Egfr is implicated in numerous disease pathologies and distinct Egfr-associated disease etiologies are known to be tissue-specific. The molecular basis of this tissue-specificity remains poorly understood. Most studies of Egfr signaling to date have been performed in vitro or in tissue-specific mouse models of disease, which has limited insight into Egfr signaling patterns in healthy tissues. Here, we carried out integrated phosphoproteomic, proteomic, and transcriptomic analyses of signaling changes across various mouse tissues in response to short-term stimulation with the Egfr ligand Egf. We show how both baseline and Egf-stimulated signaling dynamics differ between tissues. Moreover, we propose how baseline phosphorylation and total protein levels may be associated with clinically relevant tissue-specific Egfr-associated phenotypes. Altogether, our analyses illustrate tissue-specific effects of Egf stimulation and highlight potential links between underlying tissue biology and Egfr signaling output.
Collapse
Affiliation(s)
- Beatrice W. Awasthi
- Center for Systems Biology, Department of Radiation Oncology, and Center for Cancer Research, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - João A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Deborah L. Burkhart
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Ian R. Smith
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ryan L. Collins
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Cancer Program, Broad Institute of M.I.T. and Harvard, Cambridge, MA 02115, USA
| | - J. Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Kevin M. Haigis
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
2
|
Zuazo-Gaztelu I, Lawrence D, Oikonomidi I, Marsters S, Pechuan-Jorge X, Gaspar CJ, Kan D, Segal E, Clark K, Beresini M, Braun MG, Rudolph J, Modrusan Z, Choi M, Sandoval W, Reichelt M, DeWitt DC, Kujala P, van Dijk S, Klumperman J, Ashkenazi A. A nonenzymatic dependency on inositol-requiring enzyme 1 controls cancer cell cycle progression and tumor growth. PLoS Biol 2025; 23:e3003086. [PMID: 40208872 DOI: 10.1371/journal.pbio.3003086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 02/26/2025] [Indexed: 04/12/2025] Open
Abstract
Endoplasmic-reticulum resident inositol-requiring enzyme 1α (IRE1) supports protein homeostasis via its cytoplasmic kinase-RNase module. Known cancer dependency on IRE1 entails its enzymatic activation of the transcription factor XBP1s and regulated RNA decay. We discovered that some cancer cells surprisingly require IRE1 but not its enzymatic activity. IRE1 knockdown but not enzymatic IRE1 inhibition or XBP1 disruption attenuated cell cycle progression and tumor growth. IRE1 silencing led to activation of TP53 and CDKN1A/p21 in conjunction with increased DNA damage and chromosome instability, while decreasing heterochromatin as well as DNA and histone H3K9me3 methylation. Immunoelectron microscopy detected endogenous IRE1 at the nuclear envelope. Thus, cancer cells co-opt IRE1 either enzymatically or nonenzymatically, which has significant implications for IRE1's biological role and therapeutic targeting.
Collapse
Affiliation(s)
- Iratxe Zuazo-Gaztelu
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - David Lawrence
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Ioanna Oikonomidi
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Scot Marsters
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Ximo Pechuan-Jorge
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - Catarina J Gaspar
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| | - David Kan
- Department of In Vivo Pharmacology, Genentech, Inc., South San Francisco, California, United States of America
| | - Ehud Segal
- Department of In Vivo Pharmacology, Genentech, Inc., South San Francisco, California, United States of America
| | - Kevin Clark
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California, United States of America
| | - Maureen Beresini
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, California, United States of America
| | - Marie-Gabrielle Braun
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Joachim Rudolph
- Department of Discovery Chemistry, Genentech, Inc., South San Francisco, California, United States of America
| | - Zora Modrusan
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, California, United States of America
| | - Meena Choi
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, California, United States of America
| | - Wendy Sandoval
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, California, United States of America
| | - Mike Reichelt
- Department of Pathology, Genentech, Inc., South San Francisco, California, United States of America
| | - David C DeWitt
- Department of Pathology, Genentech, Inc., South San Francisco, California, United States of America
| | - Pekka Kujala
- Center for Molecular Medicine-Cell Biology, University Medical Center, Utrecht, The Netherlands
| | - Suzanne van Dijk
- Center for Molecular Medicine-Cell Biology, University Medical Center, Utrecht, The Netherlands
| | - Judith Klumperman
- Center for Molecular Medicine-Cell Biology, University Medical Center, Utrecht, The Netherlands
| | - Avi Ashkenazi
- Department of Research Oncology, Genentech, Inc., South San Francisco, California, United States of America
| |
Collapse
|
3
|
Petrova B, Guler AT. Recent Developments in Single-Cell Metabolomics by Mass Spectrometry─A Perspective. J Proteome Res 2025; 24:1493-1518. [PMID: 39437423 PMCID: PMC11976873 DOI: 10.1021/acs.jproteome.4c00646] [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: 07/28/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Recent advancements in single-cell (sc) resolution analyses, particularly in sc transcriptomics and sc proteomics, have revolutionized our ability to probe and understand cellular heterogeneity. The study of metabolism through small molecules, metabolomics, provides an additional level of information otherwise unattainable by transcriptomics or proteomics by shedding light on the metabolic pathways that translate gene expression into functional outcomes. Metabolic heterogeneity, critical in health and disease, impacts developmental outcomes, disease progression, and treatment responses. However, dedicated approaches probing the sc metabolome have not reached the maturity of other sc omics technologies. Over the past decade, innovations in sc metabolomics have addressed some of the practical limitations, including cell isolation, signal sensitivity, and throughput. To fully exploit their potential in biological research, however, remaining challenges must be thoroughly addressed. Additionally, integrating sc metabolomics with orthogonal sc techniques will be required to validate relevant results and gain systems-level understanding. This perspective offers a broad-stroke overview of recent mass spectrometry (MS)-based sc metabolomics advancements, focusing on ongoing challenges from a biologist's viewpoint, aimed at addressing pertinent and innovative biological questions. Additionally, we emphasize the use of orthogonal approaches and showcase biological systems that these sophisticated methodologies are apt to explore.
Collapse
Affiliation(s)
- Boryana Petrova
- Medical
University of Vienna, Vienna 1090, Austria
- Department
of Pathology, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
| | - Arzu Tugce Guler
- Department
of Pathology, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Institute
for Experiential AI, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
4
|
Arias-Gaguancela O, Palii C, Nissa MU, Brand M, Ranish J. QuickProt: A bioinformatics and visualization tool for DIA and PRM mass spectrometry-based proteomics datasets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645047. [PMID: 40196523 PMCID: PMC11974799 DOI: 10.1101/2025.03.24.645047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Mass spectrometry (MS)-based proteomics focuses on identifying and quantifying peptides and proteins in biological samples. Processing of MS-derived raw data, including deconvolution, alignment, and peptide-protein prediction, has been achieved through various software platforms. However, the downstream analysis, including quality control, visualizations, and interpretation of proteomics results remains challenging due to the lack of integrated tools to facilitate the analyses. To address this challenge, we developed QuickProt, a series of Python-based Google Colab notebooks for analyzing data-independent acquisition (DIA) and parallel reaction monitoring (PRM) proteomics datasets. These pipelines are designed so that users with no coding expertise can utilize the tool. Furthermore, as open-source code, QuickProt notebooks can be customized and incorporated into existing workflows. As proof of concept, we applied QuickProt to analyze in-house DIA and stable isotope dilution (SID)-PRM MS proteomics datasets from a time-course study of human erythropoiesis. The analysis resulted in annotated tables and publication-ready figures revealing a dynamic rearrangement of the proteome during erythroid differentiation, with the abundance of proteins linked to gene regulation, metabolic, and chromatin remodeling pathways increasing early in erythropoiesis. Altogether, these tools aim to automate and streamline DIA and PRM-MS proteomics data analysis, making it more efficient and less time-consuming.
Collapse
Affiliation(s)
| | - Carmen Palii
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Marjorie Brand
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Jeff Ranish
- Institute for Systems Biology, Seattle, WA, USA
| |
Collapse
|
5
|
Schneider M, Zolg DP, Samaras P, Ben Fredj S, Bold D, Guevende A, Hogrebe A, Berger MT, Graber M, Sukumar V, Mamisashvili L, Bronsthein I, Eljagh L, Gessulat S, Seefried F, Schmidt T, Frejno M. A Scalable, Web-Based Platform for Proteomics Data Processing, Result Storage and Analysis. J Proteome Res 2025; 24:1241-1249. [PMID: 39982847 PMCID: PMC11894649 DOI: 10.1021/acs.jproteome.4c00871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 02/23/2025]
Abstract
The exponential increase in proteomics data presents critical challenges for conventional processing workflows. These pipelines often consist of fragmented software packages, glued together using complex in-house scripts or error-prone manual workflows running on local hardware, which are costly to maintain and scale. The MSAID Platform offers a fully automated, managed proteomics data pipeline, consolidating formerly disjointed functions into unified, API-driven services that cover the entire process from raw data to biological insights. Backed by the cloud-native search algorithm CHIMERYS, as well as scalable cloud compute instances and data lakes, the platform facilitates efficient processing of large data sets, automation of processing via the command line, systematic result storage, analysis, and visualization. The data lake supports elastically growing storage and unified query capabilities, facilitating large-scale analyses and efficient reuse of previously processed data, such as aggregating longitudinally acquired studies. Users interact with the platform via a web interface, CLI client, or API, providing flexible, automated access. Readily available tools for accessing result data include browser-based interrogation and one-click visualizations for statistical analysis. The platform streamlines research processes, making advanced and automated proteomic workflows accessible to a broader range of scientists. The MSAID Platform is globally available via https://platform.msaid.io.
Collapse
|
6
|
Nisar M, Soman SP, Sreelan S, John L, Pinto SM, Kandasamy RK, Subbannayya Y, Prasad TSK, Kanekar S, Raju R, Devasahayam Arokia Balaya R. ProteoArk: A One-Pot Proteomics Data Analysis and Visualization Tool for Biologists. J Proteome Res 2025; 24:1008-1016. [PMID: 39928856 DOI: 10.1021/acs.jproteome.4c00556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
ProteoArk is a web-based tool that offers a range of computational pipelines for comprehensive analysis and visualization of mass spectrometry-based proteomics data. The application comprises four primary sections designed to address various aspects of mass spectrometry data analysis in a single platform, including label-free and labeled samples (SILAC/iTRAQ/TMT), differential expression analysis, and data visualization. ProteoArk supports postprocessing of Proteome Discoverer, MaxQuant, and MSFragger search results. The tool also includes functional enrichment analyses such as gene ontology, protein-protein interactions, pathway analysis, and differential expression analysis, which incorporate various statistical tests. By streamlining workflows and developing user-friendly interfaces, we created a robust and accessible solution for users with basic bioinformatics skills in proteomic data analysis. Users can easily create manuscript-ready figures with a single click, including principal component analysis, heatmaps (K-means and hierarchical), MA plots, volcano plots, and circular bar plots. ProteoArk is developed using the Django framework and is freely available for users [https://ciods.in/proteoark/]. Users can also download and run the standalone version of ProteoArk using Docker as described in the instructions [https://ciods.in/proteoark/dockerpage]. The application code, input data, and documentation are available online at https://github.com/ArokiaRex/proteoark. A tutorial video is available on YouTube: https://www.youtube.com/watch?v=WFMKAZ9Slq4&ab_channel=RexD.A.B.
Collapse
Affiliation(s)
- Mahammad Nisar
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Sreelakshmi Pathappillil Soman
- Centre for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Sourav Sreelan
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Levin John
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Sneha M Pinto
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, GU2 7XH Guildford, U.K
| | - Richard Kumaran Kandasamy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Immunology, Mayo Clinic, Rochester, Minnesota 55905, United States
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota 55905, United States
| | - Yashwanth Subbannayya
- School of Biosciences, Faculty of Health and Medical Sciences, University of Surrey, GU2 7XH Guildford, U.K
| | - Thottethodi Subrahmanya Keshava Prasad
- Centre for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Saptami Kanekar
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Rex Devasahayam Arokia Balaya
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| |
Collapse
|
7
|
Staniak M, Huang T, Figueroa-Navedo AM, Kohler D, Choi M, Hinkle T, Kleinheinz T, Blake R, Rose CM, Xu Y, Jean Beltran PM, Xue L, Bogdan M, Vitek O. Relative quantification of proteins and post-translational modifications in proteomic experiments with shared peptides: a weight-based approach. BIOINFORMATICS (OXFORD, ENGLAND) 2025; 41:btaf046. [PMID: 39888862 DOI: 10.1093/bioinformatics/btaf046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/27/2024] [Accepted: 01/28/2025] [Indexed: 02/02/2025]
Abstract
MOTIVATION Bottom-up mass spectrometry-based proteomics studies changes in protein abundance and structure across conditions. Since the currency of these experiments are peptides, i.e. subsets of protein sequences that carry the quantitative information, conclusions at a different level must be computationally inferred. The inference is particularly challenging in situations where the peptides are shared by multiple proteins or post-translational modifications. While many approaches infer the underlying abundances from unique peptides, there is a need to distinguish the quantitative patterns when peptides are shared. RESULTS We propose a statistical approach for estimating protein abundances, as well as site occupancies of post-translational modifications, based on quantitative information from shared peptides. The approach treats the quantitative patterns of shared peptides as convex combinations of abundances of individual proteins or modification sites, and estimates the abundance of each source in a sample together with the weights of the combination. In simulation-based evaluations, the proposed approach improved the precision of estimated fold changes between conditions. We further demonstrated the practical utility of the approach in experiments with diverse biological objectives, ranging from protein degradation and thermal proteome stability, to changes in protein post-translational modifications. AVAILABILITY AND IMPLEMENTATION The approach is implemented in an open-source R package MSstatsWeightedSummary. The package is currently available at https://github.com/Vitek-Lab/MSstatsWeightedSummary (doi: 10.5281/zenodo.14662989). Code required to reproduce the results presented in this article can be found in a repository https://github.com/mstaniak/MWS_reproduction (doi: 10.5281/zenodo.14656053).
Collapse
Affiliation(s)
- Mateusz Staniak
- Faculty of Mathematics and Computer Science, University of Wrocław, Wrocław, 50-383, Poland
- Centre for Statistics, Hasselt University, Diepenbeek, 3590, Belgium
| | - Ting Huang
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, United States
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, United States
| | - Amanda M Figueroa-Navedo
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, United States
| | - Devon Kohler
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, United States
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, United States
| | - Meena Choi
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Trent Hinkle
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Tracy Kleinheinz
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Robert Blake
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Christopher M Rose
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Yingrong Xu
- Discovery Sciences, Pfizer Inc., Groton, CT, 06340, United States
| | - Pierre M Jean Beltran
- Machine Learning and Computational Sciences, Pfizer Inc., Cambridge, MA, 02139, United States
| | - Liang Xue
- Machine Learning and Computational Sciences, Pfizer Inc., Cambridge, MA, 02139, United States
| | - Małgorzata Bogdan
- Faculty of Mathematics and Computer Science, University of Wrocław, Wrocław, 50-383, Poland
| | - Olga Vitek
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, United States
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, United States
| |
Collapse
|
8
|
Gonzalez-Lozano MA, Schmid EW, Whelan EM, Jiang Y, Paulo JA, Walter JC, Harper JW. EndoMAP.v1, a Structural Protein Complex Landscape of Human Endosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.636106. [PMID: 39975243 PMCID: PMC11839024 DOI: 10.1101/2025.02.07.636106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Early/sorting endosomes are dynamic organelles that play key roles in proteome control by triaging plasma membrane proteins for either recycling or degradation in the lysosome1,2,3. These events are coordinated by numerous transiently-associated regulatory complexes and integral membrane components that contribute to organelle identity during endosome maturation4. While a subset of the several hundred protein components and cargoes known to associate with endosomes have been studied at the biochemical and/or structural level, interaction partners and higher order molecular assemblies for many endosomal components remain unknown. Here, we combine cross-linking and native gel mass spectrometry5-8 of purified early endosomes with AlphaFold9,10 and computational analysis to create a systematic human endosomal structural interactome. We present dozens of structural models for endosomal protein pairs and higher order assemblies supported by experimental cross-links from their native subcellular context, suggesting structural mechanisms for previously reported regulatory processes. Using induced neurons, we validate two candidate complexes whose interactions are supported by crosslinks and structural predictions: TMEM230 as a subunit of ATP8/11 lipid flippases11 and TMEM9/9B as subunits of CLCN3/4/5 chloride-proton antiporters12. This resource and its accompanying structural network viewer provide an experimental framework for understanding organellar structural interactomes and large-scale validation of structural predictions.
Collapse
Affiliation(s)
- Miguel A Gonzalez-Lozano
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Ernst W Schmid
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA, USA
| | - Enya Miguel Whelan
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Yizhi Jiang
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Initiative in Trafficking and Neurogeneration, Department of Cell Biology, Harvard Medical School, Boston MA, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
| | - Johannes C Walter
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston MA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
9
|
Wolski WE, Grossmann J, Schwarz L, Leary P, Türker C, Nanni P, Schlapbach R, Panse C. prolfquapp ─ A User-Friendly Command-Line Tool Simplifying Differential Expression Analysis in Quantitative Proteomics. J Proteome Res 2025; 24:955-965. [PMID: 39849819 PMCID: PMC11812002 DOI: 10.1021/acs.jproteome.4c00911] [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: 10/10/2024] [Revised: 12/09/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025]
Abstract
Mass spectrometry is a cornerstone of quantitative proteomics, enabling relative protein quantification and differential expression analysis (DEA) of proteins. As experiments grow in complexity, involving more samples, groups, and identified proteins, interactive differential expression analysis tools become impractical. The prolfquapp addresses this challenge by providing a command-line interface that simplifies DEA, making it accessible to nonprogrammers and seamlessly integrating it into workflow management systems. Prolfquapp streamlines data processing and result visualization by generating dynamic HTML reports that facilitate the exploration of differential expression results. These reports allow for investigating complex experiments, such as those involving repeated measurements or multiple explanatory variables. Additionally, prolfquapp supports various output formats, including XLSX files, SummarizedExperiment objects and rank files, for further interactive analysis using spreadsheet software, the exploreDE Shiny application, or gene set enrichment analysis software, respectively. By leveraging advanced statistical models from the prolfqua R package, prolfquapp offers a user-friendly, integrated solution for large-scale quantitative proteomics studies, combining efficient data processing with insightful, publication-ready outputs.
Collapse
Affiliation(s)
- Witold E. Wolski
- Functional
Genomics Center Zurich (FGCZ) - University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
- Swiss
Institute of Bioinformatics (SIB) Quartier Sorge - Batiment Amphipole, 1015 Lausanne, Switzerland
| | - Jonas Grossmann
- Functional
Genomics Center Zurich (FGCZ) - University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
- Swiss
Institute of Bioinformatics (SIB) Quartier Sorge - Batiment Amphipole, 1015 Lausanne, Switzerland
| | - Leonardo Schwarz
- Functional
Genomics Center Zurich (FGCZ) - University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
- Swiss
Institute of Bioinformatics (SIB) Quartier Sorge - Batiment Amphipole, 1015 Lausanne, Switzerland
| | - Peter Leary
- Functional
Genomics Center Zurich (FGCZ) - University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
- Swiss
Institute of Bioinformatics (SIB) Quartier Sorge - Batiment Amphipole, 1015 Lausanne, Switzerland
| | - Can Türker
- Functional
Genomics Center Zurich (FGCZ) - University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Paolo Nanni
- Functional
Genomics Center Zurich (FGCZ) - University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Ralph Schlapbach
- Functional
Genomics Center Zurich (FGCZ) - University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Christian Panse
- Functional
Genomics Center Zurich (FGCZ) - University of Zurich/ETH Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
- Swiss
Institute of Bioinformatics (SIB) Quartier Sorge - Batiment Amphipole, 1015 Lausanne, Switzerland
| |
Collapse
|
10
|
Smith HE, Abughazaleh N, Seerattan RA, Syed F, Young D, Dufour A, Hart DA, Reimer RA, Herzog W. Sex-specific response of intramuscular fat to diet-induced obesity in rats. Sci Rep 2025; 15:2147. [PMID: 39820480 PMCID: PMC11739556 DOI: 10.1038/s41598-024-85084-7] [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: 08/22/2024] [Accepted: 12/31/2024] [Indexed: 01/19/2025] Open
Abstract
Metabolic abnormalities associated with excess adiposity in obesity contribute to many noncommunicable diseases, including sarcopenic obesity. Sarcopenic obesity is the loss of muscle mass coupled with excess fat mass and fatty infiltrations in muscle tissue called myosteatosis. A diet-induced obesity model was developed to study fat infiltration in muscle tissue. Only male rats have been considered in these investigations neglecting that female rats might respond differently. The objective of this study was to determine if the response to diet-induced obesity can be generalized to both sexes, or whether sex affects the response to the HFS diet, as indicated by markers of metabolic syndrome and changed in muscle integrity. Using a combination of histological staining techniques, quantitative proteomics, and measures of metabolic syndrome and inflammation, it was determined that the diet-induced obesity model in female Sprague-Dawley rats is a viable model with pronounced effects on the musculoskeletal system. We found sex-dependent and muscle-specific differences in intramuscular fat infiltration between male and female rats receiving the obesogenic diet. Including females in research may allow for identifying distinct causes of the mechanistic relationship between diet, obesity, metabolic syndrome, and the sex-dependent differential effects of these factors on adaptation and degeneration of musculoskeletal tissues.
Collapse
Affiliation(s)
- Hannah E Smith
- Human Performance Lab, University of Calgary, Calgary, AB, Canada.
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.
| | - Nada Abughazaleh
- Human Performance Lab, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Ruth A Seerattan
- Human Performance Lab, University of Calgary, Calgary, AB, Canada
| | - Faizan Syed
- Human Performance Lab, University of Calgary, Calgary, AB, Canada
| | - Daniel Young
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - David A Hart
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Raylene A Reimer
- Human Performance Lab, University of Calgary, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada
| | - Walter Herzog
- Human Performance Lab, University of Calgary, Calgary, AB, Canada.
- McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
11
|
Chatterjee P, Consoli CE, Schiller H, Winter KK, McCallum ME, Schulze S, Pohlschroder M. Quorum sensing mediates morphology and motility transitions in the model archaeon Haloferax volcanii. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.633064. [PMID: 39868331 PMCID: PMC11760428 DOI: 10.1101/2025.01.14.633064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Quorum sensing (QS) is a mechanism of intercellular communication that enables microbes to alter gene expression and adapt to the environment. This cell-cell signaling is necessary for intra- and interspecies behaviors such as virulence and biofilm formation. While QS has been extensively studied in bacteria, little is known about cell-cell communication in archaea. Here we established an archaeal model system to study QS. We showed that for Haloferax volcanii, the transition from motile rods to non-motile disks is dependent on a possibly novel, secreted small molecule present in cell-free conditioned medium (CM). Moreover, we determined that this putative QS molecule fails to induce the morphology transition in mutants lacking the regulatory factors, DdfA and CirA. Using quantitative proteomics of wild-type cells, we detected significant differential abundances of 236 proteins in the presence of CM. Conversely, in the ΔddfA mutant, addition of CM resulted in only 110 proteins of significant differential abundances. These results confirm that DdfA is involved in CM-dependent regulation. CirA, along with other proteins involved in morphology and swimming motility transitions, is among the proteins regulated by DdfA. These discoveries significantly advance our understanding of microbial communication within archaeal species.
Collapse
Affiliation(s)
- Priyanka Chatterjee
- University of Pennsylvania, Department of Biology, Philadelphia, PA 19104, USA
- University of Pennsylvania, Perelman School of Medicine, Department of Microbiology, Philadelphia PA 19104, USA
| | - Caroline E. Consoli
- University of Pennsylvania, Department of Chemistry, Philadelphia, PA 19104, USA
| | - Heather Schiller
- University of Pennsylvania, Department of Biology, Philadelphia, PA 19104, USA
| | - Kiersten K. Winter
- Rochester Institute of Technology, College of Science, Gosnell School of Life Sciences, NY 14623, USA
| | - Monica E. McCallum
- University of Pennsylvania, Department of Chemistry, Philadelphia, PA 19104, USA
| | - Stefan Schulze
- University of Pennsylvania, Department of Biology, Philadelphia, PA 19104, USA
- Rochester Institute of Technology, College of Science, Gosnell School of Life Sciences, NY 14623, USA
| | | |
Collapse
|
12
|
Zaccarelli A, Mattina B, Pont L, Benavente F, Zanotti I, Cioffi F, Elviri L. Synergy of Analytical Characterization and Biocompatible Extractions for the Enhancement of High-Quality Biorefinery Products from Medicago sativa. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:938-953. [PMID: 39723940 DOI: 10.1021/acs.jafc.4c09161] [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: 12/28/2024]
Abstract
This study presents the development of an analytical characterization strategy tailored to end products derived from an alfalfa (Medicago sativa)-based biorefinery with particular emphasis on protein concentrates and phenolic-enriched fractions. Our approach began with a comprehensive full-factorial experimental design aimed at optimizing the extraction process, taking care to design a biocompatible extraction protocol. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) techniques were used to characterize the molecular profile of the extracts. In particular, the extracts showed a significant relative abundance of flavonoids and isoflavonoids in both their aglycone and glycosylated forms, in which antioxidant activity was evaluated. In addition, we elucidated the proteomic profiles of the protein concentrates. This proteomic characterization served as a valuable resource for understanding the differences between these end products, providing insights that can guide informed decisions about their potential applications.
Collapse
Affiliation(s)
| | - Beatrice Mattina
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Laura Pont
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA·UB), University of Barcelona, 08028 Barcelona, Spain
- Serra Húnter Program, Generalitat de Catalunya, 08007 Barcelona, Spain
| | - Fernando Benavente
- Department of Chemical Engineering and Analytical Chemistry, Institute for Research on Nutrition and Food Safety (INSA·UB), University of Barcelona, 08028 Barcelona, Spain
| | - Ilaria Zanotti
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| | - Flavio Cioffi
- Contento Trade Srl, Pozzuolo de Friuli, 33050 Friuli-Venezia Giulia, Italy
| | - Lisa Elviri
- Department of Food and Drug, University of Parma, 43124 Parma, Italy
| |
Collapse
|
13
|
Li K, Teo GC, Yang KL, Yu F, Nesvizhskii AI. diaTracer enables spectrum-centric analysis of diaPASEF proteomics data. Nat Commun 2025; 16:95. [PMID: 39747075 PMCID: PMC11696033 DOI: 10.1038/s41467-024-55448-8] [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: 05/20/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
Data-independent acquisition has become a widely used strategy for peptide and protein quantification in liquid chromatography-tandem mass spectrometry-based proteomics studies. The integration of ion mobility separation into data-independent acquisition analysis, such as the diaPASEF technology available on Bruker's timsTOF platform, further improves the quantification accuracy and protein depth achievable using data-independent acquisition. We introduce diaTracer, a spectrum-centric computational tool optimized for diaPASEF data. diaTracer performs three-dimensional (mass to charge ratio, retention time, ion mobility) peak tracing and feature detection to generate precursor-resolved "pseudo-tandem mass spectra", facilitating direct ("spectral-library free") peptide identification and quantification from diaPASEF data. diaTracer is available as a stand-alone tool and is fully integrated into the widely used FragPipe computational platform. We demonstrate the performance of diaTracer and FragPipe using diaPASEF data from triple-negative breast cancer, cerebrospinal fluid, and plasma samples, data from phosphoproteomics and human leukocyte antigens immunopeptidomics experiments, and low-input data from a spatial proteomics study. We also show that diaTracer enables unrestricted identification of post-translational modifications from diaPASEF data using open/mass-offset searches.
Collapse
Affiliation(s)
- Kai Li
- Gilbert S. Omenn Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Guo Ci Teo
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Kevin L Yang
- Gilbert S. Omenn Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| | - Alexey I Nesvizhskii
- Gilbert S. Omenn Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA.
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
14
|
Pakkir Shah AK, Walter A, Ottosson F, Russo F, Navarro-Diaz M, Boldt J, Kalinski JCJ, Kontou EE, Elofson J, Polyzois A, González-Marín C, Farrell S, Aggerbeck MR, Pruksatrakul T, Chan N, Wang Y, Pöchhacker M, Brungs C, Cámara B, Caraballo-Rodríguez AM, Cumsille A, de Oliveira F, Dührkop K, El Abiead Y, Geibel C, Graves LG, Hansen M, Heuckeroth S, Knoblauch S, Kostenko A, Kuijpers MCM, Mildau K, Papadopoulos Lambidis S, Portal Gomes PW, Schramm T, Steuer-Lodd K, Stincone P, Tayyab S, Vitale GA, Wagner BC, Xing S, Yazzie MT, Zuffa S, de Kruijff M, Beemelmanns C, Link H, Mayer C, van der Hooft JJJ, Damiani T, Pluskal T, Dorrestein P, Stanstrup J, Schmid R, Wang M, Aron A, Ernst M, Petras D. Statistical analysis of feature-based molecular networking results from non-targeted metabolomics data. Nat Protoc 2025; 20:92-162. [PMID: 39304763 DOI: 10.1038/s41596-024-01046-3] [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: 10/31/2023] [Accepted: 07/02/2024] [Indexed: 09/22/2024]
Abstract
Feature-based molecular networking (FBMN) is a popular analysis approach for liquid chromatography-tandem mass spectrometry-based non-targeted metabolomics data. While processing liquid chromatography-tandem mass spectrometry data through FBMN is fairly streamlined, downstream data handling and statistical interrogation are often a key bottleneck. Especially users new to statistical analysis struggle to effectively handle and analyze complex data matrices. Here we provide a comprehensive guide for the statistical analysis of FBMN results, focusing on the downstream analysis of the FBMN output table. We explain the data structure and principles of data cleanup and normalization, as well as uni- and multivariate statistical analysis of FBMN results. We provide explanations and code in two scripting languages (R and Python) as well as the QIIME2 framework for all protocol steps, from data clean-up to statistical analysis. All code is shared in the form of Jupyter Notebooks ( https://github.com/Functional-Metabolomics-Lab/FBMN-STATS ). Additionally, the protocol is accompanied by a web application with a graphical user interface ( https://fbmn-statsguide.gnps2.org/ ) to lower the barrier of entry for new users and for educational purposes. Finally, we also show users how to integrate their statistical results into the molecular network using the Cytoscape visualization tool. Throughout the protocol, we use a previously published environmental metabolomics dataset for demonstration purposes. Together, the protocol, code and web application provide a complete guide and toolbox for FBMN data integration, cleanup and advanced statistical analysis, enabling new users to uncover molecular insights from their non-targeted metabolomics data. Our protocol is tailored for the seamless analysis of FBMN results from Global Natural Products Social Molecular Networking and can be easily adapted to other mass spectrometry feature detection, annotation and networking tools.
Collapse
Affiliation(s)
- Abzer K Pakkir Shah
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Axel Walter
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
- Applied Bioinformatics, Department of Computer Science, University of Tübingen, Tübingen, Germany
| | - Filip Ottosson
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen S, Denmark
| | - Francesco Russo
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen S, Denmark
| | - Marcelo Navarro-Diaz
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Judith Boldt
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- German Center for Infection Research, Partner Site Braunschweig-Hannover, Braunschweig, Germany
| | - Jarmo-Charles J Kalinski
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Department of Biochemistry and Microbiology, Rhodes University, Makhanda, South Africa
| | - Eftychia Eva Kontou
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- The Novo Nordisk Foundation for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - James Elofson
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, USA
| | - Alexandros Polyzois
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Carolina González-Marín
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Universidad EAFIT, Medellín, Antioquia, Colombia
| | - Shane Farrell
- Bigelow Laboratory for Ocean Sciences, East Boothbay, ME, USA
- School of Marine Sciences, Darling Marine Center, University of Maine, Walpole, ME, USA
| | - Marie R Aggerbeck
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | - Thapanee Pruksatrakul
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand
| | - Nathan Chan
- Department of Computer Science, University of California Riverside, Riverside, CA, USA
| | - Yunshu Wang
- Department of Computer Science, University of California Riverside, Riverside, CA, USA
| | - Magdalena Pöchhacker
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Department of Food Chemistry and Toxicology, University of Vienna, Vienna, Austria
| | - Corinna Brungs
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Beatriz Cámara
- Laboratorio de Microbiología Molecular y Biotecnología Ambiental, Centro de Biotecnología DAL, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | | | - Andres Cumsille
- Laboratorio de Microbiología Molecular y Biotecnología Ambiental, Centro de Biotecnología DAL, Universidad Técnica Federico Santa María, Valparaíso, Chile
| | - Fernanda de Oliveira
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Department of Biotechnology, Engineering School of Lorena, University of São Paulo, Lorena, São Paulo, Brazil
| | - Kai Dührkop
- Department of Bioinformatics, University of Jena, Jena, Germany
| | - Yasin El Abiead
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Christian Geibel
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Lana G Graves
- Department of Environmental Systems Analysis, University of Tübingen, Tübingen, Germany
- Leibniz Institute of Freshwater Ecology and Inland Fisheries, Berlin, Germany
| | - Martin Hansen
- Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | - Steffen Heuckeroth
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Simon Knoblauch
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Anastasiia Kostenko
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, USA
| | - Mirte C M Kuijpers
- Department of Ecology, Behavior and Evolution, University of California San Diego, San Diego, CA, USA
| | - Kevin Mildau
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Department of Analytical Chemistry, University of Vienna, Vienna, Austria
- Bioinformatics Group, Wageningen University and Research, Wageningen, the Netherlands
| | | | - Paulo Wender Portal Gomes
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Tilman Schramm
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
- Department of Biochemistry, University of California Riverside, Riverside, CA, USA
| | - Karoline Steuer-Lodd
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
- Department of Biochemistry, University of California Riverside, Riverside, CA, USA
| | - Paolo Stincone
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Sibgha Tayyab
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Giovanni Andrea Vitale
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Berenike C Wagner
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Shipei Xing
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Marquis T Yazzie
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, USA
| | - Simone Zuffa
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
- Collaborative Mass Spectrometry Innovation Center, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Martinus de Kruijff
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, Germany
| | - Christine Beemelmanns
- Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarbrücken, Germany
- Saarland University, Saarbrücken, Germany
| | - Hannes Link
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Christoph Mayer
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany
| | - Justin J J van der Hooft
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Bioinformatics Group, Wageningen University and Research, Wageningen, the Netherlands
- Department of Biochemistry, University of Johannesburg, Johannesburg, South Africa
| | - Tito Damiani
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Tomáš Pluskal
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Pieter Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA, USA
| | - Jan Stanstrup
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Frederiksberg C, Denmark
| | - Robin Schmid
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Mingxun Wang
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Department of Computer Science, University of California Riverside, Riverside, CA, USA
| | - Allegra Aron
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA
- Department of Chemistry and Biochemistry, University of Denver, Denver, CO, USA
| | - Madeleine Ernst
- Section for Clinical Mass Spectrometry, Danish Center for Neonatal Screening, Department of Congenital Disorders, Statens Serum Institut, Copenhagen S, Denmark.
| | - Daniel Petras
- Virtual Multi-Omics Laboratory, The Internet, Riverside, CA, USA.
- University of Tübingen, Interfaculty Institute of Microbiology and Infection Medicine, Tübingen, Germany.
- Department of Biochemistry, University of California Riverside, Riverside, CA, USA.
| |
Collapse
|
15
|
Ariyasinghe NR, Gupta D, Escopete S, Rai D, Stotland A, Sundararaman N, Ngu B, Dabke K, McCarthy L, Santos RS, McCain ML, Sareen D, Parker SJ. Identification of Disease-Relevant, Sex-Based Proteomic Differences in iPSC-Derived Vascular Smooth Muscle Cells. Int J Mol Sci 2024; 26:187. [PMID: 39796045 PMCID: PMC11719605 DOI: 10.3390/ijms26010187] [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: 11/24/2024] [Accepted: 12/12/2024] [Indexed: 01/13/2025] Open
Abstract
The prevalence of cardiovascular disease varies with sex, and the impact of intrinsic sex-based differences on vasculature is not well understood. Animal models can provide important insights into some aspects of human biology; however, not all discoveries in animal systems translate well to humans. To explore the impact of chromosomal sex on proteomic phenotypes, we used iPSC-derived vascular smooth muscle cells from healthy donors of both sexes to identify sex-based proteomic differences and their possible effects on cardiovascular pathophysiology. Our analysis confirmed that differentiated cells have a proteomic profile more similar to healthy primary aortic smooth muscle cells than iPSCs. We also identified sex-based differences in iPSC-derived vascular smooth muscle cells in pathways related to ATP binding, glycogen metabolic process, and cadherin binding as well as multiple proteins relevant to cardiovascular pathophysiology and disease. Additionally, we explored the role of autosomal and sex chromosomes in protein regulation, identifying that proteins on autosomal chromosomes also show sex-based regulation that may affect the protein expression of proteins from autosomal chromosomes. This work supports the biological relevance of iPSC-derived vascular smooth muscle cells as a model for disease, and further exploration of the pathways identified here can lead to the discovery of sex-specific pharmacological targets for cardiovascular disease.
Collapse
Affiliation(s)
- Nethika R. Ariyasinghe
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Divya Gupta
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Sean Escopete
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Deepika Rai
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Aleksandr Stotland
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Niveda Sundararaman
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Benjamin Ngu
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90007, USA; (B.N.); (M.L.M.)
| | - Kruttika Dabke
- Center for Bioinformatics and Functional Genomics, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
| | - Liam McCarthy
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
| | - Roberta S. Santos
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (R.S.S.); (D.S.)
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, West Hollywood, CA 90069, USA
| | - Megan L. McCain
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90007, USA; (B.N.); (M.L.M.)
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Dhruv Sareen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (R.S.S.); (D.S.)
- Cedars-Sinai Biomanufacturing Center, Cedars-Sinai Medical Center, West Hollywood, CA 90069, USA
- iPSC Core, David and Janet Polak Foundation Stem Cell Core Laboratory, Cedars-Sinai Medical Center, West Hollywood, CA 90069, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sarah J. Parker
- Advanced Clinical Biosystems Research Institute, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (N.R.A.); (D.G.); (S.E.); (D.R.); (A.S.); (N.S.); (L.M.)
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Board of Governors Innovation Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
16
|
Fang H, Shih MC, Jiang L, da Veiga Leprevost F, Jian R, Chan J, Nesvizhskii AI, Snyder MP, Tang H. Improving design and normalization of multiplex proteomics study. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.05.627093. [PMID: 39713300 PMCID: PMC11661083 DOI: 10.1101/2024.12.05.627093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Advances in multiplex mass spectrometry-based technologies have enabled high-throughput, quantitative proteome profiling of large cohort. However, certain experimental design configurations can amplify sample variability and introduce systematic biases. To address these challenges, we incorporated two novel features in a recent proteogenomic investigation: (1) the inclusion of two reference samples within each mass spectrometry run to serve as internal standards, and (2) the analysis of each specimen as technical replicates across two distinct mass spectrometry runs. Building on these enhancements, we present ProMix, a flexible analytical framework designed to fully leverage these supplementary experimental components. Using both simulated and real-world datasets, we demonstrate the improved performance of ProMix and highlight the advantages conferred by these refined experimental design strategies.
Collapse
|
17
|
Green HD, Van Horn GT, Williams T, Eberly A, Morales GH, Mann R, Hauter IM, Hadjifrangiskou M, Schmitz JE. Intra-strain colony biofilm heterogeneity in uropathogenic Escherichia coli and the effect of the NlpI lipoprotein. Biofilm 2024; 8:100214. [PMID: 39184815 PMCID: PMC11344014 DOI: 10.1016/j.bioflm.2024.100214] [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: 03/19/2024] [Revised: 06/25/2024] [Accepted: 07/09/2024] [Indexed: 08/27/2024] Open
Abstract
Biofilm growth facilitates the interaction of uropathogenic Escherichia coli (UPEC) with the host environment. The extracellular polymeric substances (EPS) of UPEC biofilms are composed prominently of curli amyloid fiber and cellulose polysaccharide. When the organism is propagated as a colony biofilm on agar media, these macromolecules can generate pronounced macroscopic structures. Moreover, curli/cellulose associate tightly with Congo red, generating a characteristic pink-to-red staining pattern when the media is supplemented with this dye. Among different clinical isolates of UPEC, changes in the abundance of curli/cellulose can lead to diverse colony biofilm phenotypes on a strain-by-strain basis. Nevertheless, for any given isolate, these phenotypes are classically homogenous throughout the colony biofilm. Here, we report that a subset of clinical UPEC isolates display heterogenous 'peppermint' colony biofilms, with distinct pale and red subpopulations. Through isolation of these subpopulations and whole genome sequencing, we demonstrate various emergent mutations associated with the phenomenon, including within the gene encoding the outer membrane lipoprotein nlpI. Deletion of nlpI within independent strain-backgrounds increased biofilm rugosity, while its overexpression induced the peppermint phenotype. Upregulation of EPS-associated proteins and transcripts was likewise observed in the absence of nlpI. Overall, these results demonstrate that EPS elaboration in UPEC is impacted by nlpI. More broadly, this phenomenon of intra-strain colony biofilm heterogeneity may be leveraged as a tool to identify additional members within the broad collection of genes that regulate or otherwise affect biofilm formation.
Collapse
Affiliation(s)
- Hamilton D. Green
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Gerald T. Van Horn
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Center for Personalized Microbiology, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Timothy Williams
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Allison Eberly
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Grace H. Morales
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Robert Mann
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Indiana M. Hauter
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Maria Hadjifrangiskou
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology & Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Personalized Microbiology, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan E. Schmitz
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology & Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Center for Personalized Microbiology, Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
18
|
Li K, Teo GC, Yang KL, Yu F, Nesvizhskii AI. diaTracer enables spectrum-centric analysis of diaPASEF proteomics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.25.595875. [PMID: 38854051 PMCID: PMC11160675 DOI: 10.1101/2024.05.25.595875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Data-independent acquisition (DIA) has become a widely used strategy for peptide and protein quantification in mass spectrometry-based proteomics studies. The integration of ion mobility separation into DIA analysis, such as the diaPASEF technology available on Bruker's timsTOF platform, further improves the quantification accuracy and protein depth achievable using DIA. We introduce diaTracer, a new spectrum-centric computational tool optimized for diaPASEF data. diaTracer performs three-dimensional (m/z, retention time, ion mobility) peak tracing and feature detection to generate precursor-resolved "pseudo-MS/MS" spectra, facilitating direct ("spectral-library free") peptide identification and quantification from diaPASEF data. diaTracer is available as a stand-alone tool and is fully integrated into the widely used FragPipe computational platform. We demonstrate the performance of diaTracer and FragPipe using diaPASEF data from triple-negative breast cancer (TNBC), cerebrospinal fluid (CSF), and plasma samples, data from phosphoproteomics and HLA immunopeptidomics experiments, and low-input data from a spatial proteomics study. We also show that diaTracer enables unrestricted identification of post-translational modifications from diaPASEF data using open/mass-offset searches.
Collapse
Affiliation(s)
- Kai Li
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Guo Ci Teo
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Kevin L. Yang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Alexey I. Nesvizhskii
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Benak D, Holzerova K, Kolar F, Chalupova M, Hlavackova M. Unveiling the proteome of the fasting heart: Insights into HIF-1 pathway regulation. Front Physiol 2024; 15:1462014. [PMID: 39469441 PMCID: PMC11513464 DOI: 10.3389/fphys.2024.1462014] [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: 07/09/2024] [Accepted: 09/30/2024] [Indexed: 10/30/2024] Open
Abstract
Fasting is a common dietary intervention known for its protective effects against metabolic and cardiovascular diseases. While its effects are mostly systemic, understanding tissue-specific changes in the heart is crucial for the identification of the mechanisms underlying fasting-induced cardioprotection. In this study, we performed a proteomic analysis of the fasting heart and attempted to clarify the molecular basis of fasting-induced cardioprotection. Our investigation identified a total of 4,652 proteins, with 127 exhibiting downregulation and 118 showing upregulation after fasting. Annotation analysis highlighted significant changes in processes such as lipid metabolism, the peroxisome pathway, and reactive oxygen species metabolism. Notably, the HIF-1 signaling pathway emerged as one of the focal points, with various HIF-1 targets exhibiting differential responses to fasting. Further experiments demonstrated downregulation of HIF-1α at both transcript and protein levels. Intriguingly, while gene expression of Egln3 decreased, its protein product PHD3 remained unaffected by fasting. The unchanged levels of pro-inflammatory cytokines indicated that the observed reduction in Hif1a expression did not stem from a decrease in basal inflammation. These findings underscore the complex regulation of the well-established cardioprotective HIF-1 signaling within the heart during 3-day fasting.
Collapse
Affiliation(s)
- Daniel Benak
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Kristyna Holzerova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Frantisek Kolar
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| | - Miloslava Chalupova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
- Department of Physiology, Faculty of Science, Charles University, Prague, Czechia
| | - Marketa Hlavackova
- Laboratory of Developmental Cardiology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
20
|
Kohler D, Staniak M, Yu F, Nesvizhskii AI, Vitek O. An MSstats workflow for detecting differentially abundant proteins in large-scale data-independent acquisition mass spectrometry experiments with FragPipe processing. Nat Protoc 2024; 19:2915-2938. [PMID: 38769142 DOI: 10.1038/s41596-024-01000-3] [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: 09/28/2023] [Accepted: 03/11/2024] [Indexed: 05/22/2024]
Abstract
Technological advances in mass spectrometry and proteomics have made it possible to perform larger-scale and more-complex experiments. The volume and complexity of the resulting data create major challenges for downstream analysis. In particular, next-generation data-independent acquisition (DIA) experiments enable wider proteome coverage than more traditional targeted approaches but require computational workflows that can manage much larger datasets and identify peptide sequences from complex and overlapping spectral features. Data-processing tools such as FragPipe, DIA-NN and Spectronaut have undergone substantial improvements to process spectral features in a reasonable time. Statistical analysis tools are needed to draw meaningful comparisons between experimental samples, but these tools were also originally designed with smaller datasets in mind. This protocol describes an updated version of MSstats that has been adapted to be compatible with large-scale DIA experiments. A very large DIA experiment, processed with FragPipe, is used as an example to demonstrate different MSstats workflows. The choice of workflow depends on the user's computational resources. For datasets that are too large to fit into a standard computer's memory, we demonstrate the use of MSstatsBig, a companion R package to MSstats. The protocol also highlights key decisions that have a major effect on both the results and the processing time of the analysis. The MSstats processing can be expected to take 1-3 h depending on the usage of MSstatsBig. The protocol can be run in the point-and-click graphical user interface MSstatsShiny or implemented with minimal coding expertise in R.
Collapse
Affiliation(s)
- Devon Kohler
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA
- Barnett Institute for Chemical and Biological Analysis, Northeastern University, Boston, MA, USA
| | | | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Olga Vitek
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, USA.
- Barnett Institute for Chemical and Biological Analysis, Northeastern University, Boston, MA, USA.
| |
Collapse
|
21
|
Zaed MA, Abdullah N, Tan KH, Hossain MH, Saidur R. Empowering Green Energy Storage Systems with MXene for a Sustainable Future. CHEM REC 2024; 24:e202400062. [PMID: 39318085 DOI: 10.1002/tcr.202400062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/30/2024] [Indexed: 09/26/2024]
Abstract
Green energy storage systems play a vital role in enabling a sustainable future by facilitating the efficient integration and utilization of renewable energy sources. The main problems related to two-dimensional (2D) materials are their difficult synthesis process, high cost, and bulk production, which hamper their performance. In recent years, MXenes have emerged as highly promising materials for enhancing the performance of energy storage devices due to their unique properties, including their high surface area, excellent electrical and thermal conductivity, and exceptional chemical stability. This paper presents a comprehensive scientific approach that explores the potential of MXenes for empowering green energy storage systems. Which indicates the novelty of the article. The paper reviews the latest advances in MXene synthesis techniques. Furthermore, investigates the application of MXenes in various energy storage technologies, such as lithium-ion batteries, supercapacitors, and emerging energy storage devices. The utilization of MXenes as electrodes in flexible and transparent energy storage devices is also discussed. Moreover, the paper highlights the potential of MXenes in addressing key challenges in energy storage, including enhancing energy storage capacity, improving cycling stability, and promoting fast charging and discharging rates. Additionally, industrial application and cost estimation of MXenes are explored. As the output of the work, we analyzed that HF and modified acid (LiF and HCl) are the established methods for synthesis. Due to high electrical conductivity, MXene materials are showing extraordinary results in energy storage and related applications. Making a composite hydrothermal method is one of the established methods. This scientific paper underscores the significant contributions of MXenes in advancing green energy storage systems, paving the way for a sustainable future driven by renewable energy sources. To facilitate the research, this article includes technical challenges and future recommendations for further research gaps in the topic.
Collapse
Affiliation(s)
- M A Zaed
- Research Center for Nano-Materials and Energy Technology (RCNMET), School of Engineering and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
| | - Norulsamani Abdullah
- Research Center for Nano-Materials and Energy Technology (RCNMET), School of Engineering and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
- Sunway Materials Smart Science & Engineering (SMS2E) Cluster, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
| | - K H Tan
- Research Center for Nano-Materials and Energy Technology (RCNMET), School of Engineering and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
| | - M H Hossain
- Department of Electrical and Electronic Engineering, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - R Saidur
- Research Center for Nano-Materials and Energy Technology (RCNMET), School of Engineering and Technology, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
- Sunway Materials Smart Science & Engineering (SMS2E) Cluster, Sunway University, No. 5, Jalan Universiti, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia
- School of Engineering, Lancaster University, Lancaster, LA1 4YW, UK
| |
Collapse
|
22
|
Reffai A, Hori M, Adusumilli R, Bermudez A, Bouzoubaa A, Pitteri S, Bennani Mechita M, Mallick P. A Proteomic Analysis of Nasopharyngeal Carcinoma in a Moroccan Subpopulation. Cancers (Basel) 2024; 16:3282. [PMID: 39409902 PMCID: PMC11476039 DOI: 10.3390/cancers16193282] [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: 07/22/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a distinct cancer of the head and neck that is highly prevalent in Southeast Asia and North Africa. Though an extensive analysis of environmental and genetic contributors has been performed, very little is known about the proteome of this disease. A proteomic analysis of formalin-fixed paraffin-embedded (FFPE) tissues can provide valuable information on protein expression and molecular patterns for both increasing our understanding of the disease and for biomarker discovery. To date, very few NPC proteomic studies have been performed, and none focused on patients from Morocco and North Africa. METHODS Label-free Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) was used to perform a proteomic analysis of FFPE tissue samples from a cohort of 41 NPC tumor samples of Morocco and North Africa origins. The LC-MS/MS data from this cohort were analyzed alongside 21 healthy controls using MaxQuant 2.4.2.0. A differential expression analysis was performed using the MSstats package in R. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional annotations were carried out using the DAVID bioinformatic tool. RESULTS 3341 proteins were identified across our NPC cases, revealing three main clusters and five DEPs with prognostic significance. The sex disparity of NPC was investigated from a proteomic perspective in which 59 DEPs were found between males and females, with significantly enriched terms associated with the immune response and gene expression. Furthermore, 26 DEPs were observed between patients with early and advanced stages of NPC with a significant cluster related to the immune response, implicating up-regulated DEPs such as IGHA, IGKC, and VAT1. Across both datasets, 6532 proteins were quantified between NPC patients and healthy controls. Among them, 1507 differentially expressed proteins (DEPs) were observed. GO and KEGG pathway analyses showed enriched terms of DEPs related to increased cellular activity, cell proliferation, and survival. PI3K and MAPK proteins as well as RAC1 BCL2 and PPIA were found to be overexpressed between cancer tissues and healthy controls. EBV infection was also one of the enriched pathways implicating its latent genes like LMP1 and LMP2 that activate several proteins and signaling pathways including NF-Kappa B, MAPK, and JAK-STAT pathways. CONCLUSION Our findings unveil the proteomic landscape of NPC for the first time in the Moroccan population. These studies additionally may provide a foundation for identifying potential biomarkers. Further research is still needed to help develop tools for the early diagnosis and treatment of NPC in Moroccan and North African populations.
Collapse
Affiliation(s)
- Ayman Reffai
- Intelligent Automation and BioMed Genomics Laboratory, Biology Department, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University-Tetouan, Tangier 90000, Morocco
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michelle Hori
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ravali Adusumilli
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Abel Bermudez
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | | | - Sharon Pitteri
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mohcine Bennani Mechita
- Intelligent Automation and BioMed Genomics Laboratory, Biology Department, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University-Tetouan, Tangier 90000, Morocco
| | - Parag Mallick
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
23
|
Shan X, Zhang A, Rezzonico MG, Tsai MC, Sanchez-Priego C, Zhang Y, Chen MB, Choi M, Andrade López JM, Phu L, Cramer AL, Zhang Q, Pattison JM, Rose CM, Hoogenraad CC, Jeong CG. Fully defined NGN2 neuron protocol reveals diverse signatures of neuronal maturation. CELL REPORTS METHODS 2024; 4:100858. [PMID: 39255791 PMCID: PMC11440061 DOI: 10.1016/j.crmeth.2024.100858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 04/26/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
NGN2-driven induced pluripotent stem cell (iPSC)-to-neuron conversion is a popular method for human neurological disease modeling. In this study, we present a standardized approach for generating neurons utilizing clonal, targeted-engineered iPSC lines with defined reagents. We demonstrate consistent production of excitatory neurons at scale and long-term maintenance for at least 150 days. Temporal omics, electrophysiological, and morphological profiling indicate continued maturation to postnatal-like neurons. Quantitative characterizations through transcriptomic, imaging, and functional assays reveal coordinated actions of multiple pathways that drive neuronal maturation. We also show the expression of disease-related genes in these neurons to demonstrate the relevance of our protocol for modeling neurological disorders. Finally, we demonstrate efficient generation of NGN2-integrated iPSC lines. These workflows, profiling data, and functional characterizations enable the development of reproducible human in vitro models of neurological disorders.
Collapse
Affiliation(s)
- Xiwei Shan
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Ai Zhang
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Mitchell G Rezzonico
- Department of OMNI Bioinformatics, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Ming-Chi Tsai
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | - Yingjie Zhang
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Michelle B Chen
- Department of Cellular and Tissue Genomics, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Meena Choi
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | - Lilian Phu
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Amber L Cramer
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Qiao Zhang
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Jillian M Pattison
- Advanced Cell Engineering, Department of Molecular Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Christopher M Rose
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Casper C Hoogenraad
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Claire G Jeong
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
24
|
Schauer SP, Cho CH, Novikova G, Roth GA, Lee J, Sharma AD, Foley AR, Ng C, Shen P, Choi M, Ma TP, Phu L, Budayeva HG, Cheung TK, Lalehzadeh G, Imperio J, Ngu H, Etxeberria A, Liang Y, Rezzonico MG, Dourado M, Huang K, Lai Z, Hokom M, Pandya NJ, Newton D, Abdel‐Haleem AM, Chan P, Lee D, Tassew NG, Sangaraju D, O'Connor D, Hötzel I, Stark KL, Chou C, Foreman O, Easton A, Wildsmith KR, Sperinde G, Rose CM, Friedman BA, Fuji RN, Weimer RM, Meilandt WJ, Sadekar S, Nugent AA, Biever A. Primate cerebrospinal fluid CHI3L1 reflects brain TREM2 agonism. Alzheimers Dement 2024; 20:5861-5888. [PMID: 39090679 PMCID: PMC11497760 DOI: 10.1002/alz.13921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/22/2024] [Accepted: 03/25/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Triggering receptor expressed on myeloid cells 2 (TREM2) agonists are being clinically evaluated as disease-modifying therapeutics for Alzheimer's disease. Clinically translatable pharmacodynamic (PD) biomarkers are needed to confirm drug activity and select the appropriate therapeutic dose in clinical trials. METHODS We conducted multi-omic analyses on paired non-human primate brain and cerebrospinal fluid (CSF), and stimulation of human induced pluripotent stem cell-derived microglia cultures after TREM2 agonist treatment, followed by validation of candidate fluid PD biomarkers using immunoassays. We immunostained microglia to characterize proliferation and clustering. RESULTS We report CSF soluble TREM2 (sTREM2) and CSF chitinase-3-like protein 1 (CHI3L1/YKL-40) as PD biomarkers for the TREM2 agonist hPara.09. The respective reduction of sTREM2 and elevation of CHI3L1 in brain and CSF after TREM2 agonist treatment correlated with transient microglia proliferation and clustering. DISCUSSION CSF CHI3L1 and sTREM2 reflect microglial TREM2 agonism and can be used as clinical PD biomarkers to monitor TREM2 activity in the brain. HIGHLIGHTS CSF soluble triggering receptor expressed on myeloid cells 2 (sTREM2) reflects brain target engagement for a novel TREM2 agonist, hPara.09. CSF chitinase-3-like protein 1 reflects microglial TREM2 agonism. Both can be used as clinical fluid biomarkers to monitor TREM2 activity in brain.
Collapse
Affiliation(s)
- Stephen P. Schauer
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Chang Hoon Cho
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Gloriia Novikova
- Department of BioinformaticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Gillie A. Roth
- Department of Preclinical and Translational Pharmacokinetics and PharmacodynamicsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Julie Lee
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Anup D. Sharma
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Alejandro R. Foley
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Carl Ng
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Philip Shen
- Department of Safety Assessment PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Meena Choi
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Taylur P. Ma
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Lilian Phu
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Hanna G. Budayeva
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Tommy K. Cheung
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Guita Lalehzadeh
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Jose Imperio
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Hai Ngu
- Department of Research PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Ainhoa Etxeberria
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Yuxin Liang
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | | | - Michelle Dourado
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Kevin Huang
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Zijuan Lai
- Department of Drug Metabolism and PharmacokineticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Martha Hokom
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Nikhil J. Pandya
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Dwight Newton
- Roche InformaticsHoffmann‐La Roche, Ltd.MississaugaOntarioCanada
| | | | - Pamela Chan
- Department of Biochemical and Cellular PharmacologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Donna Lee
- Department of Safety Assessment ToxicologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Nardos G. Tassew
- Department of Safety Assessment ToxicologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Dewakar Sangaraju
- Department of Drug Metabolism and PharmacokineticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Deborah O'Connor
- Department of ChemistryManufacturing, and ControlsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Isidro Hötzel
- Department of Antibody EngineeringGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Kimberly L. Stark
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Carolina Chou
- Department of Safety Assessment Nonclinical OperationsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Oded Foreman
- Department of Research PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Amy Easton
- Department of NeuroscienceGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Kristin R. Wildsmith
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Gizette Sperinde
- Department of BioAnalytical SciencesGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Christopher M. Rose
- Department of MicrochemistryProteomics, Lipidomics, and Next Generation SequencingGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Brad A. Friedman
- Department of BioinformaticsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Reina N. Fuji
- Department of Safety Assessment PathologyGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Robby M. Weimer
- Department of Translational ImagingGenentech, Inc.South San FranciscoCaliforniaUSA
| | | | - Shraddha Sadekar
- Department of Preclinical and Translational Pharmacokinetics and PharmacodynamicsGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Alicia A. Nugent
- Department of Human Pathobiology and OMNI Reverse TranslationGenentech, Inc.South San FranciscoCaliforniaUSA
| | - Anne Biever
- Department of Translational MedicineGenentech, Inc.South San FranciscoCaliforniaUSA
| |
Collapse
|
25
|
Reis-de-Oliveira G, Carregari VC, Sousa GRDRD, Martins-de-Souza D. OmicScope unravels systems-level insights from quantitative proteomics data. Nat Commun 2024; 15:6510. [PMID: 39095347 PMCID: PMC11297029 DOI: 10.1038/s41467-024-50875-z] [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: 02/13/2024] [Accepted: 07/13/2024] [Indexed: 08/04/2024] Open
Abstract
Shotgun proteomics analysis presents multifaceted challenges, demanding diverse tool integration for insights. Addressing this complexity, OmicScope emerges as an innovative solution for quantitative proteomics data analysis. Engineered to handle various data formats, it performs data pre-processing - including joining replicates, normalization, data imputation - and conducts differential proteomics analysis for both static and longitudinal experimental designs. Empowered by Enrichr with over 224 databases, OmicScope performs Over Representation Analysis (ORA) and Gene Set Enrichment Analysis (GSEA). Additionally, its Nebula module facilitates meta-analysis from independent datasets, providing a systems biology approach for enriched insights. Complete with a data visualization toolkit and accessible as Python package and a web application, OmicScope democratizes proteomics analysis, offering an efficient and high-quality pipeline for researchers.
Collapse
Affiliation(s)
- Guilherme Reis-de-Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
- Research Center, Boldrini Children's Hospital, Campinas, SP, Brazil.
| | - Victor Corasolla Carregari
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP, Brazil.
- University of Campinas (UNICAMP), Campinas, SP, Brazil.
- Instituto Nacional de Biomarcadores Em Neuropsiquiatria (INBION) Conselho Nacional de Desenvolvimento Científico E Tecnológico, São Paulo, Brazil.
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, Brazil.
- D'Or Institute for Research and Education (IDOR), São Paulo, Brazil.
- INCT in Modelling Human Complex Diseases with 3D Platforms (Model3D), São Paulo, Brazil.
| |
Collapse
|
26
|
Budayeva HG, Ma TP, Wang S, Choi M, Rose CM. Increasing the Throughput and Reproducibility of Activity-Based Proteome Profiling Studies with Hyperplexing and Intelligent Data Acquisition. J Proteome Res 2024; 23:2934-2947. [PMID: 38251652 PMCID: PMC11301772 DOI: 10.1021/acs.jproteome.3c00598] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/16/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Intelligent data acquisition (IDA) strategies, such as a real-time database search (RTS), have improved the depth of proteome coverage for experiments that utilize isobaric labels and gas phase purification techniques (i.e., SPS-MS3). In this work, we introduce inSeqAPI, an instrument application programing interface (iAPI) program that enables construction of novel data acquisition algorithms. First, we analyze biotinylated cysteine peptides from ABPP experiments to demonstrate that a real-time search method within inSeqAPI performs similarly to an equivalent vendor method. Then, we describe PairQuant, a method within inSeqAPI designed for the hyperplexing approach that utilizes protein-level isotopic labeling and peptide-level TMT labeling. PairQuant allows for TMT analysis of 36 conditions in a single sample and achieves ∼98% coverage of both peptide pair partners in a hyperplexed experiment as well as a 40% improvement in the number of quantified cysteine sites compared with non-RTS acquisition. We applied this method in the ABPP study of ligandable cysteine sites in the nucleus leading to an identification of additional druggable sites on protein- and DNA-interaction domains of transcription regulators and on nuclear ubiquitin ligases.
Collapse
Affiliation(s)
- Hanna G. Budayeva
- Department
of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., South
San Francisco, California 94080, United States
| | - Taylur P. Ma
- Department
of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., South
San Francisco, California 94080, United States
| | - Shuai Wang
- Department
of Metabolism and Pharmacokinetics, Genentech,
Inc., South San Francisco, California 94080, United States
| | - Meena Choi
- Department
of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., South
San Francisco, California 94080, United States
| | - Christopher M. Rose
- Department
of Microchemistry, Proteomics and Lipidomics, Genentech, Inc., South
San Francisco, California 94080, United States
| |
Collapse
|
27
|
Skawinski CLS, Shah PS. I'm Walking into Spiderwebs: Making Sense of Protein-Protein Interaction Data. J Proteome Res 2024; 23:2723-2732. [PMID: 38556766 PMCID: PMC11296932 DOI: 10.1021/acs.jproteome.3c00892] [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] [Indexed: 04/02/2024]
Abstract
Protein-protein interactions (PPIs) are at the heart of the molecular landscape permeating life. Proteomics studies can explore this protein interaction landscape using mass spectrometry (MS). Thanks to their high sensitivity, mass spectrometers can easily identify thousands of proteins within a single sample, but that same sensitivity generates tangled spiderwebs of data that hide biologically relevant findings. So, what does a researcher do when she finds herself walking into spiderwebs? In a field focused on discovery, MS data require rigor in their analysis, experimental validation, or a combination of both. In this Review, we provide a brief primer on MS-based experimental methods to identify PPIs. We discuss approaches to analyze the resulting data and remove the proteomic background. We consider the advantages between comprehensive and targeted studies. We also discuss how scoring might be improved through AI-based protein structure information. Women have been essential to the development of proteomics, so we will specifically highlight work by women that has made this field thrive in recent years.
Collapse
Affiliation(s)
| | - Priya S. Shah
- Department of Chemical Engineering, University of California – Davis, California
- Department of Microbiology and Molecular Genetics, University of California – Davis, California
| |
Collapse
|
28
|
Fröhlich K, Fahrner M, Brombacher E, Seredynska A, Maldacker M, Kreutz C, Schmidt A, Schilling O. Data-Independent Acquisition: A Milestone and Prospect in Clinical Mass Spectrometry-Based Proteomics. Mol Cell Proteomics 2024; 23:100800. [PMID: 38880244 PMCID: PMC11380018 DOI: 10.1016/j.mcpro.2024.100800] [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: 02/02/2024] [Revised: 06/08/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024] Open
Abstract
Data-independent acquisition (DIA) has revolutionized the field of mass spectrometry (MS)-based proteomics over the past few years. DIA stands out for its ability to systematically sample all peptides in a given m/z range, allowing an unbiased acquisition of proteomics data. This greatly mitigates the issue of missing values and significantly enhances quantitative accuracy, precision, and reproducibility compared to many traditional methods. This review focuses on the critical role of DIA analysis software tools, primarily focusing on their capabilities and the challenges they address in proteomic research. Advances in MS technology, such as trapped ion mobility spectrometry, or high field asymmetric waveform ion mobility spectrometry require sophisticated analysis software capable of handling the increased data complexity and exploiting the full potential of DIA. We identify and critically evaluate leading software tools in the DIA landscape, discussing their unique features, and the reliability of their quantitative and qualitative outputs. We present the biological and clinical relevance of DIA-MS and discuss crucial publications that paved the way for in-depth proteomic characterization in patient-derived specimens. Furthermore, we provide a perspective on emerging trends in clinical applications and present upcoming challenges including standardization and certification of MS-based acquisition strategies in molecular diagnostics. While we emphasize the need for continuous development of software tools to keep pace with evolving technologies, we advise researchers against uncritically accepting the results from DIA software tools. Each tool may have its own biases, and some may not be as sensitive or reliable as others. Our overarching recommendation for both researchers and clinicians is to employ multiple DIA analysis tools, utilizing orthogonal analysis approaches to enhance the robustness and reliability of their findings.
Collapse
Affiliation(s)
- Klemens Fröhlich
- Proteomics Core Facility, Biozentrum Basel, University of Basel, Basel, Switzerland
| | - Matthias Fahrner
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany
| | - Eva Brombacher
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany; Centre for Integrative Biological Signaling Studies (CIBSS), University of Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Adrianna Seredynska
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Maximilian Maldacker
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Clemens Kreutz
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany; Centre for Integrative Biological Signaling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum Basel, University of Basel, Basel, Switzerland
| | - Oliver Schilling
- Institute for Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany.
| |
Collapse
|
29
|
Webel H, Niu L, Nielsen AB, Locard-Paulet M, Mann M, Jensen LJ, Rasmussen S. Imputation of label-free quantitative mass spectrometry-based proteomics data using self-supervised deep learning. Nat Commun 2024; 15:5405. [PMID: 38926340 PMCID: PMC11208500 DOI: 10.1038/s41467-024-48711-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
Imputation techniques provide means to replace missing measurements with a value and are used in almost all downstream analysis of mass spectrometry (MS) based proteomics data using label-free quantification (LFQ). Here we demonstrate how collaborative filtering, denoising autoencoders, and variational autoencoders can impute missing values in the context of LFQ at different levels. We applied our method, proteomics imputation modeling mass spectrometry (PIMMS), to an alcohol-related liver disease (ALD) cohort with blood plasma proteomics data available for 358 individuals. Removing 20 percent of the intensities we were able to recover 15 out of 17 significant abundant protein groups using PIMMS-VAE imputations. When analyzing the full dataset we identified 30 additional proteins (+13.2%) that were significantly differentially abundant across disease stages compared to no imputation and found that some of these were predictive of ALD progression in machine learning models. We, therefore, suggest the use of deep learning approaches for imputing missing values in MS-based proteomics on larger datasets and provide workflows for these.
Collapse
Affiliation(s)
- Henry Webel
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Lili Niu
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Annelaura Bach Nielsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Marie Locard-Paulet
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Lars Juhl Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Simon Rasmussen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N, Denmark.
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| |
Collapse
|
30
|
Kochanowsky JA, Mira PM, Elikaee S, Muratore K, Rai AK, Riestra AM, Johnson PJ. Trichomonas vaginalis extracellular vesicles up-regulate and directly transfer adherence factors promoting host cell colonization. Proc Natl Acad Sci U S A 2024; 121:e2401159121. [PMID: 38865261 PMCID: PMC11194581 DOI: 10.1073/pnas.2401159121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/16/2024] [Indexed: 06/14/2024] Open
Abstract
Trichomonas vaginalis, a common sexually transmitted parasite that colonizes the human urogenital tract, secretes extracellular vesicles (TvEVs) that are taken up by human cells and are speculated to be taken up by parasites as well. While the crosstalk between TvEVs and human cells has led to insight into host:parasite interactions, roles for TvEVs in infection have largely been one-sided, with little known about the effect of TvEV uptake by T. vaginalis. Approximately 11% of infections are found to be coinfections of multiple T. vaginalis strains. Clinical isolates often differ in their adherence to and cytolysis of host cells, underscoring the importance of understanding the effects of TvEV uptake within the parasite population. To address this question, our lab tested the ability of a less adherent strain of T. vaginalis, G3, to take up fluorescently labeled TvEVs derived from both itself (G3-EVs) and TvEVs from a more adherent strain of the parasite (B7RC2-EVs). Here, we showed that TvEVs generated from the more adherent strain are internalized more efficiently compared to the less adherent strain. Additionally, preincubation of G3 parasites with B7RC2-EVs increases parasite aggregation and adherence to host cells. Transcriptomics revealed that TvEVs up-regulate expression of predicted parasite membrane proteins and identified an adherence factor, heteropolysaccharide binding protein (HPB2). Finally, using comparative proteomics and superresolution microscopy, we demonstrated direct transfer of an adherence factor, cadherin-like protein, from TvEVs to the recipient parasite's surface. This work identifies TvEVs as a mediator of parasite:parasite communication that may impact pathogenesis during mixed infections.
Collapse
Affiliation(s)
- Joshua A. Kochanowsky
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Portia M. Mira
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Samira Elikaee
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Katherine Muratore
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Anand Kumar Rai
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| | - Angelica M. Riestra
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
- Department of Biology, San Diego State University, San Diego, CA92182
| | - Patricia J. Johnson
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA90095
| |
Collapse
|
31
|
Roberts DS, Loo JA, Tsybin YO, Liu X, Wu S, Chamot-Rooke J, Agar JN, Paša-Tolić L, Smith LM, Ge Y. Top-down proteomics. NATURE REVIEWS. METHODS PRIMERS 2024; 4:38. [PMID: 39006170 PMCID: PMC11242913 DOI: 10.1038/s43586-024-00318-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 07/16/2024]
Abstract
Proteoforms, which arise from post-translational modifications, genetic polymorphisms and RNA splice variants, play a pivotal role as drivers in biology. Understanding proteoforms is essential to unravel the intricacies of biological systems and bridge the gap between genotypes and phenotypes. By analysing whole proteins without digestion, top-down proteomics (TDP) provides a holistic view of the proteome and can decipher protein function, uncover disease mechanisms and advance precision medicine. This Primer explores TDP, including the underlying principles, recent advances and an outlook on the future. The experimental section discusses instrumentation, sample preparation, intact protein separation, tandem mass spectrometry techniques and data collection. The results section looks at how to decipher raw data, visualize intact protein spectra and unravel data analysis. Additionally, proteoform identification, characterization and quantification are summarized, alongside approaches for statistical analysis. Various applications are described, including the human proteoform project and biomedical, biopharmaceutical and clinical sciences. These are complemented by discussions on measurement reproducibility, limitations and a forward-looking perspective that outlines areas where the field can advance, including potential future applications.
Collapse
Affiliation(s)
- David S Roberts
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
| | - Joseph A Loo
- Department of Chemistry and Biochemistry, Department of Biological Chemistry, University of California - Los Angeles, Los Angeles, CA, USA
| | | | - Xiaowen Liu
- Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Si Wu
- Department of Chemistry and Biochemistry, The University of Alabama, Tuscaloosa, AL, USA
| | | | - Jeffrey N Agar
- Departments of Chemistry and Chemical Biology and Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Ljiljana Paša-Tolić
- Environmental and Molecular Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lloyd M Smith
- Department of Chemistry, University of Wisconsin, Madison, WI, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin, Madison, WI, USA
- Department of Cell and Regenerative Biology, Human Proteomics Program, University of Wisconsin - Madison, Madison, WI, USA
| |
Collapse
|
32
|
Ergin EK, Myung JJ, Lange PF. Statistical Testing for Protein Equivalence Identifies Core Functional Modules Conserved across 360 Cancer Cell Lines and Presents a General Approach to Investigating Biological Systems. J Proteome Res 2024; 23:2169-2185. [PMID: 38804581 PMCID: PMC11166143 DOI: 10.1021/acs.jproteome.4c00131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/04/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
Quantitative proteomics has enhanced our capability to study protein dynamics and their involvement in disease using various techniques, including statistical testing, to discern the significant differences between conditions. While most focus is on what is different between conditions, exploring similarities can provide valuable insights. However, exploring similarities directly from the analyte level, such as proteins, genes, or metabolites, is not a standard practice and is not widely adopted. In this study, we propose a statistical framework called QuEStVar (Quantitative Exploration of Stability and Variability through statistical hypothesis testing), enabling the exploration of quantitative stability and variability of features with a combined statistical framework. QuEStVar utilizes differential and equivalence testing to expand statistical classifications of analytes when comparing conditions. We applied our method to an extensive data set of cancer cell lines and revealed a quantitatively stable core proteome across diverse tissues and cancer subtypes. The functional analysis of this set of proteins highlighted the molecular mechanism of cancer cells to maintain constant conditions of the tumorigenic environment via biological processes, including transcription, translation, and nucleocytoplasmic transport.
Collapse
Affiliation(s)
- Enes K. Ergin
- Department
of Pathology, University of British Columbia, Vancouver, British Columbia V6T 1Z7, Canada
- Michael
Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, British Columbia V5Z 2H4, Canada
| | - Junia J.K. Myung
- Department
of Pathology, University of British Columbia, Vancouver, British Columbia V6T 1Z7, Canada
- Michael
Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, British Columbia V5Z 2H4, Canada
| | - Philipp F. Lange
- Department
of Pathology, University of British Columbia, Vancouver, British Columbia V6T 1Z7, Canada
- Michael
Cuccione Childhood Cancer Research Program, BC Children’s Hospital Research Institute, Vancouver, British Columbia V5Z 2H4, Canada
| |
Collapse
|
33
|
Hollingsworth LR, Veeraraghavan P, Paulo JA, Harper JW. Spatiotemporal proteomic profiling of cellular responses to NLRP3 agonists. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590338. [PMID: 38659763 PMCID: PMC11042255 DOI: 10.1101/2024.04.19.590338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Nucleotide-binding domain and leucine-rich repeat pyrin-domain containing protein 3 (NLRP3) is an innate immune sensor that forms an inflammasome in response to various cellular stressors. Gain-of-function mutations in NLRP3 cause autoinflammatory diseases and NLRP3 signalling itself exacerbates the pathogenesis of many other human diseases. Despite considerable therapeutic interest, the primary drivers of NLRP3 activation remain controversial due to the diverse array of signals that are integrated through NLRP3. Here, we mapped subcellular proteome changes to lysosomes, mitochondrion, EEA1-positive endosomes, and Golgi caused by the NLRP3 inflammasome agonists nigericin and CL097. We identified several common disruptions to retrograde trafficking pathways, including COPI and Shiga toxin-related transport, in line with recent studies. We further characterized mouse NLRP3 trafficking throughout its activation using temporal proximity proteomics, which supports a recent model of NLRP3 recruitment to endosomes during inflammasome activation. Collectively, these findings provide additional granularity to our understanding of the molecular events driving NLRP3 activation and serve as a valuable resource for cell biological research. We have made our proteomics data accessible through an open-access Shiny browser to facilitate future research within the community, available at: https://harperlab.connect.hms.harvard.edu/inflame/. We will display anonymous peer review for this manuscript on pubpub.org (https://harperlab.pubpub.org/pub/nlrp3/) rather than a traditional journal. Moreover, we invite community feedback on the pubpub version of this manuscript, and we will address criticisms accordingly.
Collapse
Affiliation(s)
- L. Robert Hollingsworth
- Department of Cell Biology, Harvard Medical School, Harvard
University, Boston, MA 02115, USA
| | | | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Harvard
University, Boston, MA 02115, USA
| | - J. Wade Harper
- Department of Cell Biology, Harvard Medical School, Harvard
University, Boston, MA 02115, USA
| |
Collapse
|
34
|
Chasseigneaux S, Cochois-Guégan V, Lecorgne L, Lochus M, Nicolic S, Blugeon C, Jourdren L, Gomez-Zepeda D, Tenzer S, Sanquer S, Nivet-Antoine V, Menet MC, Laplanche JL, Declèves X, Cisternino S, Saubaméa B. Fasting upregulates the monocarboxylate transporter MCT1 at the rat blood-brain barrier through PPAR δ activation. Fluids Barriers CNS 2024; 21:33. [PMID: 38589879 PMCID: PMC11003008 DOI: 10.1186/s12987-024-00526-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 02/29/2024] [Indexed: 04/10/2024] Open
Abstract
BACKGROUND The blood-brain barrier (BBB) is pivotal for the maintenance of brain homeostasis and it strictly regulates the cerebral transport of a wide range of endogenous compounds and drugs. While fasting is increasingly recognized as a potential therapeutic intervention in neurology and psychiatry, its impact upon the BBB has not been studied. This study was designed to assess the global impact of fasting upon the repertoire of BBB transporters. METHODS We used a combination of in vivo and in vitro experiments to assess the response of the brain endothelium in male rats that were fed ad libitum or fasted for one to three days. Brain endothelial cells were acutely purified and transcriptionaly profiled using RNA-Seq. Isolated brain microvessels were used to assess the protein expression of selected BBB transporters through western blot. The molecular mechanisms involved in the adaptation to fasting were investigated in primary cultured rat brain endothelial cells. MCT1 activity was probed by in situ brain perfusion. RESULTS Fasting did not change the expression of the main drug efflux ATP-binding cassette transporters or P-glycoprotein activity at the BBB but modulated a restrictive set of solute carrier transporters. These included the ketone bodies transporter MCT1, which is pivotal for the brain adaptation to fasting. Our findings in vivo suggested that PPAR δ, a major lipid sensor, was selectively activated in brain endothelial cells in response to fasting. This was confirmed in vitro where pharmacological agonists and free fatty acids selectively activated PPAR δ, resulting in the upregulation of MCT1 expression. Moreover, dosing rats with a specific PPAR δ antagonist blocked the upregulation of MCT1 expression and activity induced by fasting. CONCLUSIONS Altogether, our study shows that fasting affects a selected set of BBB transporters which does not include the main drug efflux transporters. Moreover, we describe a previously unknown selective adaptive response of the brain vasculature to fasting which involves PPAR δ and is responsible for the up-regulation of MCT1 expression and activity. Our study opens new perspectives for the metabolic manipulation of the BBB in the healthy or diseased brain.
Collapse
Affiliation(s)
- Stéphanie Chasseigneaux
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Véronique Cochois-Guégan
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Lucas Lecorgne
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Murielle Lochus
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Sophie Nicolic
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Corinne Blugeon
- Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Laurent Jourdren
- Département de biologie, GenomiqueENS, Institut de Biologie de l'ENS (IBENS), École normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - David Gomez-Zepeda
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), A Hemlholtz Institute of the DKFZ, Mainz, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Division 191, 69120, Heidelberg, Germany
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Stefan Tenzer
- Helmholtz-Institute for Translational Oncology Mainz (HI-TRON Mainz), A Hemlholtz Institute of the DKFZ, Mainz, Germany
- German Cancer Research Center (DKFZ) Heidelberg, Division 191, 69120, Heidelberg, Germany
- Institute of Immunology, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | | | - Valérie Nivet-Antoine
- AP-HP Biochimie générale, Hôpital Necker Enfants Malades, Université Paris Cité, Inserm, Innovations Thérapeutiques en Hémostase, Paris, France
| | - Marie-Claude Menet
- Institut de Chimie Physique, CNRS UMR8000, Université Paris-Saclay, 91400, Orsay, France
| | - Jean-Louis Laplanche
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Xavier Declèves
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Salvatore Cisternino
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Bruno Saubaméa
- Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Cité, Inserm, 4 avenue de l'Observatoire, 75006, Paris, France.
| |
Collapse
|
35
|
Manda V, Pavelka J, Lau E. Proteomics applications in next generation induced pluripotent stem cell models. Expert Rev Proteomics 2024; 21:217-228. [PMID: 38511670 PMCID: PMC11065590 DOI: 10.1080/14789450.2024.2334033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 03/08/2024] [Indexed: 03/22/2024]
Abstract
INTRODUCTION Induced pluripotent stem (iPS) cell technology has transformed biomedical research. New opportunities now exist to create new organoids, microtissues, and body-on-a-chip systems for basic biology investigations and clinical translations. AREAS COVERED We discuss the utility of proteomics for attaining an unbiased view into protein expression changes during iPS cell differentiation, cell maturation, and tissue generation. The ability to discover cell-type specific protein markers during the differentiation and maturation of iPS-derived cells has led to new strategies to improve cell production yield and fidelity. In parallel, proteomic characterization of iPS-derived organoids is helping to realize the goal of bridging in vitro and in vivo systems. EXPERT OPINIONS We discuss some current challenges of proteomics in iPS cell research and future directions, including the integration of proteomic and transcriptomic data for systems-level analysis.
Collapse
Affiliation(s)
- Vyshnavi Manda
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Consortium for Fibrosis Research and Translation, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jay Pavelka
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Consortium for Fibrosis Research and Translation, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Edward Lau
- Department of Medicine, Division of Cardiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Consortium for Fibrosis Research and Translation, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
36
|
Williams AS, Crown SB, Lyons SP, Koves TR, Wilson RJ, Johnson JM, Slentz DH, Kelly DP, Grimsrud PA, Zhang GF, Muoio DM. Ketone flux through BDH1 supports metabolic remodeling of skeletal and cardiac muscles in response to intermittent time-restricted feeding. Cell Metab 2024; 36:422-437.e8. [PMID: 38325337 PMCID: PMC10961007 DOI: 10.1016/j.cmet.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/20/2023] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
Time-restricted feeding (TRF) has gained attention as a dietary regimen that promotes metabolic health. This study questioned if the health benefits of an intermittent TRF (iTRF) schedule require ketone flux specifically in skeletal and cardiac muscles. Notably, we found that the ketolytic enzyme beta-hydroxybutyrate dehydrogenase 1 (BDH1) is uniquely enriched in isolated mitochondria derived from heart and red/oxidative skeletal muscles, which also have high capacity for fatty acid oxidation (FAO). Using mice with BDH1 deficiency in striated muscles, we discover that this enzyme optimizes FAO efficiency and exercise tolerance during acute fasting. Additionally, iTRF leads to robust molecular remodeling of muscle tissues, and muscle BDH1 flux does indeed play an essential role in conferring the full adaptive benefits of this regimen, including increased lean mass, mitochondrial hormesis, and metabolic rerouting of pyruvate. In sum, ketone flux enhances mitochondrial bioenergetics and supports iTRF-induced remodeling of skeletal muscle and heart.
Collapse
Affiliation(s)
- Ashley S Williams
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Scott B Crown
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Scott P Lyons
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Timothy R Koves
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Geriatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Rebecca J Wilson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Jordan M Johnson
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Dorothy H Slentz
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA
| | - Daniel P Kelly
- Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Paul A Grimsrud
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA
| | - Deborah M Muoio
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, NC 27701, USA; Department of Medicine, Division of Endocrinology, Metabolism, and Nutrition, Duke University Medical Center, Durham, NC 27710, USA; Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
37
|
Alvarez-Jarreta J, Amos B, Aurrecoechea C, Bah S, Barba M, Barreto A, Basenko EY, Belnap R, Blevins A, Böhme U, Brestelli J, Brown S, Callan D, Campbell LI, Christophides GK, Crouch K, Davison HR, DeBarry JD, Demko R, Doherty R, Duan Y, Dundore W, Dyer S, Falke D, Fischer S, Gajria B, Galdi D, Giraldo-Calderón GI, Harb OS, Harper E, Helb D, Howington C, Hu S, Humphrey J, Iodice J, Jones A, Judkins J, Kelly SA, Kissinger JC, Kittur N, Kwon DK, Lamoureux K, Li W, Lodha D, MacCallum RM, Maslen G, McDowell MA, Myers J, Nural MV, Roos DS, Rund SSC, Shanmugasundram A, Sitnik V, Spruill D, Starns D, Tomko SS, Wang H, Warrenfeltz S, Wieck R, Wilkinson PA, Zheng J. VEuPathDB: the eukaryotic pathogen, vector and host bioinformatics resource center in 2023. Nucleic Acids Res 2024; 52:D808-D816. [PMID: 37953350 PMCID: PMC10767879 DOI: 10.1093/nar/gkad1003] [Citation(s) in RCA: 51] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/09/2023] [Accepted: 10/19/2023] [Indexed: 11/14/2023] Open
Abstract
The Eukaryotic Pathogen, Vector and Host Informatics Resource (VEuPathDB, https://veupathdb.org) is a Bioinformatics Resource Center funded by the National Institutes of Health with additional funding from the Wellcome Trust. VEuPathDB supports >600 organisms that comprise invertebrate vectors, eukaryotic pathogens (protists and fungi) and relevant free-living or non-pathogenic species or hosts. Since 2004, VEuPathDB has analyzed omics data from the public domain using contemporary bioinformatic workflows, including orthology predictions via OrthoMCL, and integrated the analysis results with analysis tools, visualizations, and advanced search capabilities. The unique data mining platform coupled with >3000 pre-analyzed data sets facilitates the exploration of pertinent omics data in support of hypothesis driven research. Comparisons are easily made across data sets, data types and organisms. A Galaxy workspace offers the opportunity for the analysis of private large-scale datasets and for porting to VEuPathDB for comparisons with integrated data. The MapVEu tool provides a platform for exploration of spatially resolved data such as vector surveillance and insecticide resistance monitoring. To address the growing body of omics data and advances in laboratory techniques, VEuPathDB has added several new data types, searches and features, improved the Galaxy workspace environment, redesigned the MapVEu interface and updated the infrastructure to accommodate these changes.
Collapse
Affiliation(s)
| | - Beatrice Amos
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | | | - Saikou Bah
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | | | - Ana Barreto
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Evelina Y Basenko
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | | | - Ann Blevins
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | | | | | - Stuart Brown
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | - Kathryn Crouch
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Helen R Davison
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | | | - Richard Demko
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan Doherty
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yikun Duan
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Sarah Dyer
- European Bioinformatics Institute, Hinxton CB10 1SD, UK
| | - Dave Falke
- University of Georgia, Athens, GA 30602, USA
| | - Steve Fischer
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Bindu Gajria
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Galdi
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Omar S Harb
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Danica Helb
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Sufen Hu
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - John Iodice
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew Jones
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - John Judkins
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah A Kelly
- Imperial College London, South Kensington, London SW7 2BU, UK
| | | | | | - Dae Kun Kwon
- University of Notre Dame, Notre Dame, IN 46556, USA
| | | | - Wei Li
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Disha Lodha
- European Bioinformatics Institute, Hinxton CB10 1SD, UK
| | | | - Gareth Maslen
- Imperial College London, South Kensington, London SW7 2BU, UK
| | | | - Jeremy Myers
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - David S Roos
- University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Achchuthan Shanmugasundram
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
- Genomics England Limited, London E14 5AB, UK
| | - Vasily Sitnik
- European Bioinformatics Institute, Hinxton CB10 1SD, UK
| | | | - David Starns
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | | | | | | | - Robert Wieck
- University of Notre Dame, Notre Dame, IN 46556, USA
| | - Paul A Wilkinson
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Jie Zheng
- University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
38
|
Hemm L, Miucci A, Kraus A, Riediger M, Tholen S, Abdelaziz N, Georg J, Schilling O, Hess WR. Interactors and effects of overexpressing YlxR/RnpM, a conserved RNA binding protein in cyanobacteria. RNA Biol 2024; 21:1-19. [PMID: 39625117 PMCID: PMC11622646 DOI: 10.1080/15476286.2024.2429230] [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] [Revised: 10/23/2024] [Accepted: 11/05/2024] [Indexed: 12/08/2024] Open
Abstract
Throughout the tree of life RNA-binding proteins play important roles, but they are poorly characterized in cyanobacteria. Overexpression of the predicted RNA-binding protein Ssr1238 in the cyanobacterium Synechocystis 6803 for 24 h led to higher levels of RNase P RNA, tRNAs, and stress-related mRNAs. Co-immunoprecipitation of proteins followed by MS analysis and sequencing of UV crosslinked, co-immunoprecipitated RNA samples identified potential interaction partners of Ssr1238. The most enriched transcript was RNase P RNA, and RnpA, the protein component of RNase P, was among the most highly enriched proteins. A second highly enriched transcript is derived from gene ssl3177, which encodes a central enzyme in cell wall remodelling during cell division. The data also showed a strong connection to the RNA maturation and modification system indicated by co-precipitation of RNA modifying enzymes, riboendonuclease E and enolase. Surprisingly, cyanophycin synthetase and urease were highly enriched as well. In conclusion, Ssr1238 specifically binds to two different transcripts and could be involved in the coordination of RNA maturation, translation, cell division, and aspects of nitrogen metabolism. Our results are consistent with recent findings that the B. subtilis YlxR protein functions as an RNase P modulator (RnpM), extending its proposed role to the phylum cyanobacteria, and suggesting additional functionalities.
Collapse
Affiliation(s)
- Luisa Hemm
- Genetics and Experimental Bioinformatics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Anna Miucci
- Genetics and Experimental Bioinformatics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Alexander Kraus
- Genetics and Experimental Bioinformatics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Matthias Riediger
- Genetics and Experimental Bioinformatics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Stefan Tholen
- Institute for Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nouha Abdelaziz
- Genetics and Experimental Bioinformatics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Jens Georg
- Genetics and Experimental Bioinformatics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang R. Hess
- Genetics and Experimental Bioinformatics, Faculty of Biology, University of Freiburg, Freiburg, Germany
| |
Collapse
|
39
|
van Oostrum M, Blok TM, Giandomenico SL, Tom Dieck S, Tushev G, Fürst N, Langer JD, Schuman EM. The proteomic landscape of synaptic diversity across brain regions and cell types. Cell 2023; 186:5411-5427.e23. [PMID: 37918396 PMCID: PMC10686415 DOI: 10.1016/j.cell.2023.09.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/18/2023] [Accepted: 09/28/2023] [Indexed: 11/04/2023]
Abstract
Neurons build synaptic contacts using different protein combinations that define the specificity, function, and plasticity potential of synapses; however, the diversity of synaptic proteomes remains largely unexplored. We prepared synaptosomes from 7 different transgenic mouse lines with fluorescently labeled presynaptic terminals. Combining microdissection of 5 different brain regions with fluorescent-activated synaptosome sorting (FASS), we isolated and analyzed the proteomes of 18 different synapse types. We discovered ∼1,800 unique synapse-type-enriched proteins and allocated thousands of proteins to different types of synapses (https://syndive.org/). We identify shared synaptic protein modules and highlight the proteomic hotspots for synapse specialization. We reveal unique and common features of the striatal dopaminergic proteome and discover the proteome signatures that relate to the functional properties of different interneuron classes. This study provides a molecular systems-biology analysis of synapses and a framework to integrate proteomic information for synapse subtypes of interest with cellular or circuit-level experiments.
Collapse
Affiliation(s)
- Marc van Oostrum
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Thomas M Blok
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | | | | | - Georgi Tushev
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Nicole Fürst
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany
| | - Julian D Langer
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany; Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Erin M Schuman
- Max Planck Institute for Brain Research, Frankfurt am Main, Germany.
| |
Collapse
|
40
|
Harris L, Fondrie WE, Oh S, Noble WS. Evaluating Proteomics Imputation Methods with Improved Criteria. J Proteome Res 2023; 22:3427-3438. [PMID: 37861703 PMCID: PMC10949645 DOI: 10.1021/acs.jproteome.3c00205] [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] [Indexed: 10/21/2023]
Abstract
Quantitative measurements produced by tandem mass spectrometry proteomics experiments typically contain a large proportion of missing values. Missing values hinder reproducibility, reduce statistical power, and make it difficult to compare across samples or experiments. Although many methods exist for imputing missing values, in practice, the most commonly used methods are among the worst performing. Furthermore, previous benchmarking studies have focused on relatively simple measurements of error such as the mean-squared error between imputed and held-out values. Here we evaluate the performance of commonly used imputation methods using three practical, "downstream-centric" criteria. These criteria measure the ability to identify differentially expressed peptides, generate new quantitative peptides, and improve the peptide lower limit of quantification. Our evaluation comprises several experiment types and acquisition strategies, including data-dependent and data-independent acquisition. We find that imputation does not necessarily improve the ability to identify differentially expressed peptides but that it can identify new quantitative peptides and improve the peptide lower limit of quantification. We find that MissForest is generally the best performing method per our downstream-centric criteria. We also argue that existing imputation methods do not properly account for the variance of peptide quantifications and highlight the need for methods that do.
Collapse
Affiliation(s)
- Lincoln Harris
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
| | | | - Sewoong Oh
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| | - William S Noble
- Department of Genome Sciences, University of Washington, Seattle, Washington 98195, United States
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
41
|
Jones J, MacKrell EJ, Wang TY, Lomenick B, Roukes ML, Chou TF. Tidyproteomics: an open-source R package and data object for quantitative proteomics post analysis and visualization. BMC Bioinformatics 2023; 24:239. [PMID: 37280522 DOI: 10.1186/s12859-023-05360-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/25/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND The analysis of mass spectrometry-based quantitative proteomics data can be challenging given the variety of established analysis platforms, the differences in reporting formats, and a general lack of approachable standardized post-processing analyses such as sample group statistics, quantitative variation and even data filtering. We developed tidyproteomics to facilitate basic analysis, improve data interoperability and potentially ease the integration of new processing algorithms, mainly through the use of a simplified data-object. RESULTS The R package tidyproteomics was developed as both a framework for standardizing quantitative proteomics data and a platform for analysis workflows, containing discrete functions that can be connected end-to-end, thus making it easier to define complex analyses by breaking them into small stepwise units. Additionally, as with any analysis workflow, choices made during analysis can have large impacts on the results and as such, tidyproteomics allows researchers to string each function together in any order, select from a variety of options and in some cases develop and incorporate custom algorithms. CONCLUSIONS Tidyproteomics aims to simplify data exploration from multiple platforms, provide control over individual functions and analysis order, and serve as a tool to assemble complex repeatable processing workflows in a logical flow. Datasets in tidyproteomics are easy to work with, have a structure that allows for biological annotations to be added, and come with a framework for developing additional analysis tools. The consistent data structure and accessible analysis and plotting tools also offers a way for researchers to save time on mundane data manipulation tasks.
Collapse
Affiliation(s)
- Jeff Jones
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, 91125, USA.
- Division of Physics, Mathematics and Astronomy, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA, 91125, USA.
| | - Elliot J MacKrell
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA, 91125, USA
| | - Ting-Yu Wang
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Brett Lomenick
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Michael L Roukes
- Division of Physics, Mathematics and Astronomy, California Institute of Technology, 1200 East California Boulevard, Pasadena, CA, 91125, USA
| | - Tsui-Fen Chou
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA, 91125, USA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| |
Collapse
|