1
|
Evangelista JE, Ali-Nasser T, Malek LE, Xie Z, Marino GB, Bester AC, Ma'ayan A. lncRNAlyzr: Enrichment Analysis for lncRNA Sets. J Mol Biol 2025; 437:168938. [PMID: 40133794 PMCID: PMC12145269 DOI: 10.1016/j.jmb.2025.168938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 03/27/2025]
Abstract
lncRNAs make up a large portion of the human genome affecting many biological processes in normal physiology and diseases. However, human lncRNAs are understudied compared to protein-coding genes. While there are many tools for performing gene set enrichment analysis for coding genes, few tools exist for lncRNA enrichment analysis. lncRNAlyzr is a webserver application designed for lncRNAs enrichment analysis. lncRNAlyzr has a database containing 33 lncRNA set libraries created by computing correlations between lncRNAs and annotated coding gene sets. After users submit a set of lncRNAs to lncRNAlyzr, the enrichment analysis results are visualized as ball-and-stick subnetworks where nodes are lncRNAs connected to enrichment terms from across selected lncRNA set libraries. To demonstrate lncRNAlyzr, it was used to analyze the effects of knocking down the lncRNA CYTOR in K562 cells. Overall, lncRNAlyzr is an enrichment analysis tool for lncRNAs aiming to further our understanding of lncRNAs functional modules. lncRNAlyzr is available from: https://lncrnalyzr.maayanlab.cloud.
Collapse
Affiliation(s)
- John Erol Evangelista
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Tahleel Ali-Nasser
- Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel.
| | - Lauren E Malek
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Zhuorui Xie
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Giacomo B Marino
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Assaf C Bester
- Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel.
| | - Avi Ma'ayan
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| |
Collapse
|
2
|
Chen Y, Zhao N, Xu L, Jia X, Liu F, Huang J, Li X, Wang Y, Lai C, Shen Y, Wang F, Lv Y, Huang X, Zhang F, Gu H, Dai S. Integrative multi-omics analysis reveals the LncRNA 60967.1-PLCD4-ATRA axis as a key regulator of colorectal cancer progression and immune response. Mol Cancer 2025; 24:164. [PMID: 40481569 PMCID: PMC12142938 DOI: 10.1186/s12943-025-02359-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 05/19/2025] [Indexed: 06/11/2025] Open
Abstract
Colorectal cancer (CRC) is a major global health concern, characterized by high morbidity and mortality rates. CRC progression involves intricate molecular networks that remain incompletely understood. In this study, we conducted an integrative multi-omics analysis of transcriptomic, proteomic, and metabolomic profiles from CRC tissues and matched normal adjacent tissues (NATs). Our analysis revealed 1,394 differentially expressed long non-Coding RNAs (lncRNAs), 2,788 genes, 548 proteins, and 91 metabolites. A significant interaction network comprising 22 lncRNAs, 14 mRNAs/proteins, and 9 metabolites was identified, among which lncRNA 60967.1 emerged as a pivotal regulator. Functional validation demonstrated that lncRNA 60967.1 is markedly downregulated in CRC cell lines and patient tissues. Overexpression of lncRNA 60967.1 restored expression of the tumor suppressor PLCD4 and increased levels of all-trans retinoic acid (ATRA). This modulation enhanced IFN-γ-induced apoptosis and increased expression of the IFN-γ receptor subunit IFNGR1, thereby partially reversing IFN-γ resistance. In murine models, lncRNA 60967.1 overexpression promoted immune cell infiltration and synergized with anti-PD-1 therapy to inhibit tumor growth. Collectively, our findings uncover a novel lncRNA-mRNA/protein-metabolite network, the lncRNA 60967.1-PLCD4-ATRA axis, that plays a critical role in CRC progression and immune modulation, offering promising therapeutic targets for improved treatment efficacy.
Collapse
Affiliation(s)
- Yiyi Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Ningning Zhao
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China.
| | - Lingna Xu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Xiya Jia
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Fang Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China
| | - Jian Huang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China
| | - Xuhua Li
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China
| | - Yunfei Wang
- Hangzhou ShengTing Medical Technology Co., Ltd, Hangzhou, Zhejiang Province, 310018, China
| | - Chuanxi Lai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Yanbin Shen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Fei Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Yiming Lv
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Xuefeng Huang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Fan Zhang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China.
| | - Hongcang Gu
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, Anhui Province, 230031, China.
| | - Sheng Dai
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
- Provincial Key Laboratory of Precise Diagnosis and Treatment of Abdominal Infection, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
3
|
Du Y, Wu J. A unified pan-cancer proteome atlas. Cancer Cell 2025:S1535-6108(25)00221-1. [PMID: 40513574 DOI: 10.1016/j.ccell.2025.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2025] [Revised: 05/22/2025] [Accepted: 05/22/2025] [Indexed: 06/16/2025]
Abstract
In this issue of Cancer Cell, Knol et al. present the Pan-Cancer Proteome Atlas (TPCPA), a proteomic resource developed using single-shot data-independent acquisition mass spectrometry (DIA-MS). TPCPA provides proteome-scale quantifications of 999 tumors across 22 cancer types in a unified manner, for discovering tumor biology, biomarkers, and therapeutic targets.
Collapse
Affiliation(s)
- Yang Du
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jianmin Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Center for Cancer Bioinformatics, Peking University Cancer Hospital & Institute, Beijing 100142, China; Peking University International Cancer Institute, Peking University, Beijing 100191, China.
| |
Collapse
|
4
|
Müller-Dott S, Jaehnig EJ, Munchic KP, Jiang W, Yaron-Barir TM, Savage SR, Garrido-Rodriguez M, Johnson JL, Lussana A, Petsalaki E, Lei JT, Dugourd A, Krug K, Cantley LC, Mani DR, Zhang B, Saez-Rodriguez J. Comprehensive evaluation of phosphoproteomic-based kinase activity inference. Nat Commun 2025; 16:4771. [PMID: 40404650 PMCID: PMC12098709 DOI: 10.1038/s41467-025-59779-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 05/05/2025] [Indexed: 05/24/2025] Open
Abstract
Kinases regulate cellular processes and are essential for understanding cellular function and disease. To investigate the regulatory state of a kinase, numerous methods have been developed to infer kinase activities from phosphoproteomics data using kinase-substrate libraries. However, few phosphorylation sites can be attributed to an upstream kinase in these libraries, limiting the scope of kinase activity inference. Moreover, inferred activities vary across methods, necessitating evaluation for accurate interpretation. Here, we present benchmarKIN, an R package enabling comprehensive evaluation of kinase activity inference methods. Alongside classical perturbation experiments, benchmarKIN introduces a tumor-based benchmarking approach utilizing multi-omics data to identify highly active or inactive kinases. We used benchmarKIN to evaluate kinase-substrate libraries, inference algorithms and the potential of adding predicted kinase-substrate interactions to overcome the coverage limitations. Our evaluation shows most computational methods perform similarly, but the choice of library impacts the inferred activities with a combination of manually curated libraries demonstrating superior performance in recapitulating kinase activities. Additionally, in the tumor-based evaluation, adding predicted targets from NetworKIN further boosts the performance. We then demonstrate how kinase activity inference aids characterize kinase inhibitor responses in cell lines. Overall, benchmarKIN helps researchers to select reliable methods for identifying deregulated kinases.
Collapse
Affiliation(s)
- Sophia Müller-Dott
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Eric J Jaehnig
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | | | - Wen Jiang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Tomer M Yaron-Barir
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Sara R Savage
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Martin Garrido-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Jared L Johnson
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alessandro Lussana
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridgeshire, UK
| | - Evangelia Petsalaki
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridgeshire, UK
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | - Aurelien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany
| | - Karsten Krug
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Lewis C Cantley
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - D R Mani
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, Bioquant, Heidelberg, Germany.
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridgeshire, UK.
| |
Collapse
|
5
|
Yang Y, Wang S, Chen Y, Wang X, Jiang W, Jin Y, Zeng W, Wu D, Shen B, Yang H. Ontolomics-P: Advancing Proteomics Data Interpretation through GPT-4o Reannotated Topic Ontology and Data-Driven Analysis. Anal Chem 2025; 97:10299-10308. [PMID: 40326493 DOI: 10.1021/acs.analchem.5c00390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
The interpretation of proteomics data often relies on functional enrichment analysis, such as Gene Ontology (GO) enrichment, to uncover the biological functions of proteins, as well as the examination of protein expression patterns across data sets like the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database. However, conventional approaches to functional enrichment frequently produce extensive and redundant term lists, complicating interpretation and synthesis. Moreover, the absence of specialized tools tailored to proteomics researchers limits the efficient exploration of protein expression within specific biological contexts. To address these challenges, we developed Ontolomics-P, a user-friendly web-based tool designed to advance proteomics data interpretation. Ontolomics-P integrates topic modeling using latent Dirichlet allocation (LDA) with GO semantic similarity analysis, enabling the consolidation of redundant terms into coherent topics. These topics are further refined and reannotated using the GPT-4o language model, creating a novel topics database that provides precise and interpretable insights into shared biological functions. Additionally, Ontolomics-P incorporates quantitative proteomic data from 10 diverse cancer types archived in the CPTAC database, allowing for a comprehensive exploration of protein expression profiles from a data-driven perspective. Through detailed case studies, we demonstrate the tool's capacity to streamline workflows, simplify interpretation, and provide actionable biological insights. Ontolomics-P represents a significant advancement in proteomics data analysis, offering innovative solutions for functional annotation, quantitative exploration, and visualization, ultimately empowering researchers to accelerate discoveries in systems biology and beyond.
Collapse
Affiliation(s)
- Yin Yang
- Liver Surgery and NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu 610041, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shisheng Wang
- Liver Surgery and NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu 610041, China
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuzhe Chen
- Liver Surgery and NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinyuan Wang
- Proteomics and Metabolomics Core Facilities, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wei Jiang
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youmei Jin
- Proteomics and Metabolomics Core Facilities, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenjuan Zeng
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dongbo Wu
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Disease, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bairong Shen
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao Yang
- Liver Surgery and NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medical Research Center, Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu 610041, China
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
- Proteomics and Metabolomics Core Facilities, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
6
|
Heller EM, Barthel K, Räschle M, Schukken KM, Sheltzer JM, Storchová Z. Explainable Machine Learning Identifies Factors for Dosage Compensation in Aneuploid Human Cancer Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.12.653427. [PMID: 40463217 PMCID: PMC12132375 DOI: 10.1101/2025.05.12.653427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2025]
Abstract
Aneuploidy, a hallmark of cancer, leads to widespread changes in chromosome copy number, altering the abundance of hundreds or thousands of proteins. How-ever, evidence suggests that levels of proteins encoded on affected chromosomes are often buffered toward their abundances observed in diploid cells. Despite its preval-ence, the molecular mechanisms driving this protein dosage compensation remain largely unknown. It is unclear whether all proteins are buffered to a similar degree, what factors determine buffering, and whether dosage compensation varies across different cell lines or tumor types. Moreover, its potential adaptive advantage and therapeutic relevance remain unexplored. Here, we established a novel approach to quantify protein dosage buffering in a gene copy number-dependent manner, show-ing that dosage compensation is widespread but variable in cancer cell lines and in vivo tumor samples. By developing multifactorial machine learning models, we identify mean gene dependency, protein complex participation, haploinsufficiency, and mRNA decay as key predictors of buffering. We also show that dosage com-pensation can affect oncogenic potential and that higher buffering correlates with reduced proteotoxic stress and increased drug resistance. These findings highlight protein dosage compensation as a crucial regulatory mechanism and a potential therapeutic target in aneuploid cancers.
Collapse
Affiliation(s)
- Erik Marcel Heller
- Department of Molecular Genetics, Faculty of Biology, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Karen Barthel
- Department of Molecular Genetics, Faculty of Biology, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Markus Räschle
- Department of Molecular Genetics, Faculty of Biology, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Kaiserslautern, Germany
| | - Klaske M. Schukken
- Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Jason M. Sheltzer
- Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Zuzana Storchová
- Department of Molecular Genetics, Faculty of Biology, Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Kaiserslautern, Germany
| |
Collapse
|
7
|
Xu M, Xu B. Protein lipidation in the tumor microenvironment: enzymology, signaling pathways, and therapeutics. Mol Cancer 2025; 24:138. [PMID: 40335986 PMCID: PMC12057185 DOI: 10.1186/s12943-025-02309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/18/2025] [Indexed: 05/09/2025] Open
Abstract
Protein lipidation is a pivotal post-translational modification that increases protein hydrophobicity and influences their function, localization, and interaction network. Emerging evidence has shown significant roles of lipidation in the tumor microenvironment (TME). However, a comprehensive review of this topic is lacking. In this review, we present an integrated and in-depth literature review of protein lipidation in the context of the TME. Specifically, we focus on three major lipidation modifications: S-prenylation, S-palmitoylation, and N-myristoylation. We emphasize how these modifications affect oncogenic signaling pathways and the complex interplay between tumor cells and the surrounding stromal and immune cells. Furthermore, we explore the therapeutic potential of targeting lipidation mechanisms in cancer treatment and discuss prospects for developing novel anticancer strategies that disrupt lipidation-dependent signaling pathways. By bridging protein lipidation with the dynamics of the TME, our review provides novel insights into the complex relationship between them that drives tumor initiation and progression.
Collapse
Affiliation(s)
- Mengke Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China.
| |
Collapse
|
8
|
Bartha Á, Weltz B, Betancourt LH, Gil J, Pinto de Almeida N, Bianchini G, Szeitz B, Szadai L, Pla I, Kemény LV, Jánosi ÁJ, Hong R, Rajeh A, Nogueira F, Doma V, Woldmar N, Guedes J, Újfaludi Z, Kim Y, Szarvas T, Pahi Z, Pankotai T, Szasz AM, Sanchez A, Baldetorp B, Tímár J, Németh IB, Kárpáti S, Appelqvist R, Domont GB, Pawlowski K, Wieslander E, Malm J, Fenyo D, Horvatovich P, Marko-Varga G, Győrffy B. Melanoma Proteomics Unveiled: Harmonizing Diverse Data Sets for Biomarker Discovery and Clinical Insights via MEL-PLOT. J Proteome Res 2025. [PMID: 40322912 DOI: 10.1021/acs.jproteome.4c00749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Using several melanoma proteomics data sets we created a single analysis platform that enables the discovery, knowledge build, and validation of diagnostic, predictive, and prognostic biomarkers at the protein level. Quantitative mass-spectrometry-based proteomic data was obtained from five independent cohorts, including 489 tissue samples from 394 patients with accompanying clinical metadata. We established an interactive R-based web platform that enables the comparison of protein levels across diverse cohorts, and supports correlation analysis between proteins and clinical metadata including survival outcomes. By comparing differential protein levels between metastatic, primary tumor, and nonmalignant samples in two of the cohorts, we identified 274 proteins showing significant differences among the sample types. Further analysis of these 274 proteins in lymph node metastatic samples from a third cohort revealed that 45 proteins exhibited a significant effect on patient survival. The three most significant proteins were HP (HR = 4.67, p = 2.8e-06), LGALS7 (HR = 3.83, p = 2.9e-05), and UBQLN1 (HR = 3.2, p = 4.8e-05). The user-friendly interactive web platform, accessible at https://www.tnmplot.com/melanoma, provides an interactive interface for the analysis of proteomic and clinical data. The MEL-PLOT platform, through its interactive capabilities, streamlines the creation of a comprehensive knowledge base, empowering hypothesis formulation and diligent monitoring of the most recent advancements in the domains of biomedical research and drug development.
Collapse
Affiliation(s)
- Áron Bartha
- Department of Bioinformatics, Semmelweis University, Budapest 1085, Hungary
- Department of Pediatrics, Semmelweis University, Budapest 1085, Hungary
| | - Boglárka Weltz
- Department of Bioinformatics, Semmelweis University, Budapest 1085, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, H-1117, Budapest, Hungary
| | - Lazaro Hiram Betancourt
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | - Jeovanis Gil
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | - Natália Pinto de Almeida
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | | | - Beáta Szeitz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, Budapest, 1085, Hungary
| | - Leticia Szadai
- Department of Dermatology and Allergology, University of Szeged, Szeged, 6720, Hungary
| | - Indira Pla
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Department of Biomedical Engineering, Faculty of Engineering, LTH, Lund University, Lund, 22363, Sweden
| | - Lajos V Kemény
- HCEMM-SU Translational Dermatology Research Group, Semmelweis University, Budapest, 1085, Hungary
- Department of Physiology, Semmelweis University, Budapest, 1094, Hungary
- Department of Dermatology, Venerology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, 1085, Hungary
| | - Ágnes Judit Jánosi
- Department of Dermatology and Allergology, University of Szeged, Szeged, 6720, Hungary
| | - Runyu Hong
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, New York 10016, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Ahmad Rajeh
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Fábio Nogueira
- Proteomics Unit, Institute of Chemistry and Research Center for Precision Medicine, Institute of Biophysics Carlos Chagas Filho, Federal Univesity of Rio de Janeiro, Rio de Janeiro, 21941-170, Brazil
| | - Viktória Doma
- Department of Dermatology, Venerology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, 1085, Hungary
| | - Nicole Woldmar
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Chemistry Institute Federal, University of Rio de Janeiro, Rio de Janiero, 21941-909, Brazil
| | - Jéssica Guedes
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
- Chemistry Institute Federal, University of Rio de Janeiro, Rio de Janiero, 21941-909, Brazil
| | - Zsuzsanna Újfaludi
- University of Szeged, Albert Szent-Györgyi Clinical Centre, Department of Pathology, 6720, Szeged, Hungary
| | - Yonghyo Kim
- Drug Discovery Platform Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Tibor Szarvas
- Department of Urology, Semmelweis University, Budapest, 1082, Hungary
- Department of Urology, University of Duisburg-Essen, Essen, 45147, Germany
| | - Zoltan Pahi
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged H-6725, Hungary
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core Group, University of Szeged, Budapesti út 9, Szeged H-6728, Hungary
| | - Tibor Pankotai
- Department of Pathology, Albert Szent-Györgyi Medical School, University of Szeged, Állomás utca 1, Szeged H-6725, Hungary
- Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Genome Integrity and DNA Repair Core Group, University of Szeged, Budapesti út 9, Szeged H-6728, Hungary
- Competence Centre of the Life Sciences Cluster of the Centre of Excellence for Interdisciplinary Research, Development and Innovation, University of Szeged, Dugonics tér 13, Szeged H-6720, Hungary
| | - A Marcell Szasz
- Division of Oncology, Department of Internal Medicine and Oncology, Semmelweis University, 1085 Budapest, Hungary
| | - Aniel Sanchez
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö,205 02, Sweden
| | - Bo Baldetorp
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund,221 84, Sweden
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Faculty of Medicine, Semmelweis University, Budapest,1085, Hungary
| | - István Balázs Németh
- Department of Dermatology and Allergology, University of Szeged, Szeged, 6720, Hungary
| | - Sarolta Kárpáti
- Department of Dermatology, Venerology and Dermatooncology, Faculty of Medicine, Semmelweis University, Budapest, 1085, Hungary
| | - Roger Appelqvist
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | - Gilberto Barbosa Domont
- Proteomics Unit, Institute of Chemistry and Research Center for Precision Medicine, Institute of Biophysics Carlos Chagas Filho, Federal Univesity of Rio de Janeiro, Rio de Janeiro, 21941-170, Brazil
| | - Krzysztof Pawlowski
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö,205 02, Sweden
- Department of Biochemistry and Microbiology, Warsaw University of Life Sciences, Warszawa,02-787, Poland
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9148, United States
| | - Elisabet Wieslander
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Skåne University Hospital Malmö, Malmö,205 02, Sweden
| | - Johan Malm
- Section for Clinical Chemistry, Department of Translational Medicine, Lund University, Lund 21428, Sweden
| | - David Fenyo
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, New York 10016, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Peter Horvatovich
- University of Groningen, Groningen Research Institute of Pharmacy, Analytical Biochemistry, Groningen, 9711, The Netherlands
| | - György Marko-Varga
- European Cancer Moonshot Lund Center, Lund, SE-221 84, Sweden
- Clinical Protein Science & Imaging, Biomedical Centre, Department of Biomedical Engineering, Lund University, Lund, 223 63, Sweden
| | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, Budapest 1085, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, H-1117, Budapest, Hungary
- Dept. of Biophysics, Medical School, University of Pecs, H-7624, Pecs, Hungary
| |
Collapse
|
9
|
Wang H, Zhou R, Xu C, Dai L, Hou R, Zheng L, Fu C, Shi G, Wang J, Li Y, Cen J, Xu X, Yu L, Li Y, Wang J, Du Q, Li Z. GRP78 Nanobody-Directed Immunotoxin Activates Innate Immunity Through STING Pathway to Synergize Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408086. [PMID: 40135833 PMCID: PMC12097070 DOI: 10.1002/advs.202408086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 02/27/2025] [Indexed: 03/27/2025]
Abstract
The lack of targetable antigens poses a significant challenge in developing effective cancer-targeted therapies. Cell surface translocation of endoplasmic reticulum (ER) chaperones, such as glucose-regulated protein 78 (GRP78), during malignancy, drug resistance, and ER stress induced by therapies, offers a promising pan-cancer target. To target GRP78, nanobody C5, identified from a phage library and exhibiting high affinity for human and mouse GRP78, is utilized to develop the Pseudomonas exotoxin (PE) immunotoxin C5-PE38. C5-PE38 induced ER stress, apoptosis and immunogenic cell death in targeted cells and showed antitumor efficacy against colorectal cancer and melanoma models without obvious toxicity. Mechanistically, transcriptome profiling showed that C5-PE38 reshaped the tumor immune microenvironment with enhanced innate and adaptive immune response and response to interferon beta. Moreover, C5-PE38-induced cell death could trans-activate STING pathway in dendritic cells and macrophages, promoting CD8+ T cell infiltration. It also sensitizes both primary and metastatic melanomas to anti-PD1 therapy, partly through STING activation. Overall, this study unveils a feasible GRP78 nanobody-directed therapy strategy for single or combinatorial cancer intervention. This work finds that C5-PE38-induced cell death stimulates STING-dependent cytosolic DNA release to promote antitumor immunity, a mechanism not previously reported for PE38, providing valuable insights for its clinical use.
Collapse
Affiliation(s)
- Huifang Wang
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Post‐doctoral Scientific Research Station of Basic MedicineJinan UniversityGuangzhou510632China
| | - Runhua Zhou
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Chengchao Xu
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Lingyun Dai
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Rui Hou
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Harry Perkins Institute of Medical ResearchQEII Medical Centre and Centre for Medical ResearchThe University of Western AustraliaNedlandsWA6009Australia
| | - Liuhai Zheng
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Chunjin Fu
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Guangwei Shi
- Department of Neurosurgery & Medical Research CenterShunde HospitalSouthern Medical University (The First People's Hospital of Shunde Foshan)Guangzhou510515China
| | - Jingwei Wang
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yang Li
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Jinpeng Cen
- Department of UrologyNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xiaolong Xu
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Le Yu
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
- School of Traditional Chinese Medicine and School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yilei Li
- Clinical Pharmacy CenterNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jigang Wang
- School of Traditional Chinese Medicine and School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di HerbsArtemisinin Research CenterInstitute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700China
- State Key Laboratory of Antiviral DrugsSchool of PharmacyHenan UniversityKaifeng475004China
| | - Qingfeng Du
- School of Traditional Chinese Medicine and School of Pharmaceutical SciencesGuangdong Provincial Key Laboratory of New Drug ScreeningSchool of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zhijie Li
- Department of Critical Care MedicineGuangdong Provincial Clinical Research Center for GeriatricsShenzhen Clinical Research Centre for GeriatricsDepartment of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| |
Collapse
|
10
|
Martins Rodrigues F, Terekhanova NV, Imbach KJ, Clauser KR, Esai Selvan M, Mendizabal I, Geffen Y, Akiyama Y, Maynard M, Yaron TM, Li Y, Cao S, Storrs EP, Gonda OS, Gaite-Reguero A, Govindan A, Kawaler EA, Wyczalkowski MA, Klein RJ, Turhan B, Krug K, Mani DR, Leprevost FDV, Nesvizhskii AI, Carr SA, Fenyö D, Gillette MA, Colaprico A, Iavarone A, Robles AI, Huang KL, Kumar-Sinha C, Aguet F, Lazar AJ, Cantley LC, Marigorta UM, Gümüş ZH, Bailey MH, Getz G, Porta-Pardo E, Ding L. Precision proteogenomics reveals pan-cancer impact of germline variants. Cell 2025; 188:2312-2335.e26. [PMID: 40233739 DOI: 10.1016/j.cell.2025.03.026] [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: 10/09/2023] [Revised: 04/29/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
We investigate the impact of germline variants on cancer patients' proteomes, encompassing 1,064 individuals across 10 cancer types. We introduced an approach, "precision peptidomics," mapping 337,469 coding germline variants onto peptides from patients' mass spectrometry data, revealing their potential impact on post-translational modifications, protein stability, allele-specific expression, and protein structure by leveraging the relevant protein databases. We identified rare pathogenic and common germline variants in cancer genes potentially affecting proteomic features, including variants altering protein abundance and structure and variants in kinases (ERBB2 and MAP2K2) impacting phosphorylation. Precision peptidome analysis predicted destabilizing events in signal-regulatory protein alpha (SIRPA) and glial fibrillary acid protein (GFAP), relevant to immunomodulation and glioblastoma diagnostics, respectively. Genome-wide association studies identified quantitative trait loci for gene expression and protein levels, spanning millions of SNPs and thousands of proteins. Polygenic risk scores correlated with distal effects from risk variants. Our findings emphasize the contribution of germline genetics to cancer heterogeneity and high-throughput precision peptidomics.
Collapse
Affiliation(s)
- Fernanda Martins Rodrigues
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Nadezhda V Terekhanova
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kathleen J Imbach
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isabel Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Translational Prostate Cancer Research Lab, CIC bioGUNE-Basurto, Biocruces Bizkaia Health Research Institute, Derio, Spain
| | - Yifat Geffen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Yo Akiyama
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Tomer M Yaron
- Meyer Cancer Center, Department of Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Erik P Storrs
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Olivia S Gonda
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA
| | - Adrian Gaite-Reguero
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Akshay Govindan
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Emily A Kawaler
- Applied Bioinformatics Laboratories, New York University Langone Health, New York City, NY, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Berk Turhan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karsten Krug
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - D R Mani
- Broad Institute of MIT and Harvard, Cambridge, MA, 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
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Antonio Colaprico
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Antonio Iavarone
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD, USA
| | - Kuan-Lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Alexander J Lazar
- Departments of Pathology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Urko M Marigorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Matthew H Bailey
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA.
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Eduard Porta-Pardo
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Barcelona Supercomputing Center (BSC), Barcelona, Spain.
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, Saint Louis, MO, USA.
| |
Collapse
|
11
|
Campeanu IJ, Jiang Y, Afisllari H, Dzinic S, Polin L, Yang ZQ. Multi-omics analysis reveals CMTR1 upregulation in cancer and roles in ribosomal protein gene expression and tumor growth. Cell Commun Signal 2025; 23:197. [PMID: 40275371 PMCID: PMC12023683 DOI: 10.1186/s12964-025-02147-6] [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: 11/21/2024] [Accepted: 03/09/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND CMTR1 (cap methyltransferase 1), a key nuclear mRNA cap methyltransferase, catalyzes 2'-O-methylation of the first transcribed nucleotide, a critical step in mRNA cap formation. Previous studies have implicated CMTR1 in embryonic stem cell differentiation and immune responses during viral infection; however, its role in cancer biology remains largely unexplored. This study aims to elucidate CMTR1's function in cancer progression and evaluate its potential as a novel therapeutic target in certain cancer types. METHODS We conducted a comprehensive multi-omics analysis of CMTR1 across various human cancers using TCGA and CPTAC datasets. Functional studies were performed using CRISPR-mediated knockout and siRNA knockdown in human and mouse basal-like breast cancer models. Transcriptomic and pathway enrichment analyses were carried out in CMTR1 knockout/knockdown models to identify CMTR1-regulated genes. In silico screening and biochemical assays were employed to identify novel CMTR1 inhibitors. RESULTS Multi-omics analysis revealed that CMTR1 is significantly upregulated at the mRNA, protein, and phosphoprotein levels across multiple cancer types in the TCGA and CPTAC datasets. Functional studies demonstrated that CMTR1 depletion significantly inhibits tumor growth both in vitro and in vivo. Transcriptomic analysis of CMTR1 knockout cells revealed that CMTR1 primarily regulates ribosomal protein genes and other transcripts containing 5' Terminal Oligopyrimidine (TOP) motifs. Additionally, CMTR1 affects the expression of snoRNA host genes and snoRNAs, suggesting a broader role in RNA metabolism. Mechanistic studies indicated that CMTR1's target specificity is partly determined by mRNA structure, particularly the presence of 5'TOP motifs. Finally, through in silico screening and biochemical assays, we identified several novel CMTR1 inhibitors, including N97911, which demonstrated in vitro growth inhibition activity in breast cancer cells. CONCLUSIONS Our findings establish CMTR1 as an important player in cancer biology, regulating critical aspects of RNA metabolism and ribosome biogenesis. The study highlights CMTR1's potential as a therapeutic target in certain cancer types and provides a foundation for developing novel cancer treatments targeting mRNA cap methylation.
Collapse
Affiliation(s)
- Ion John Campeanu
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yuanyuan Jiang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Hilda Afisllari
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Sijana Dzinic
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Lisa Polin
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Zeng-Quan Yang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA.
| |
Collapse
|
12
|
Kołodziejczak-Guglas I, Simões RLS, de Souza Santos E, Demicco EG, Lazcano Segura RN, Ma W, Wang P, Geffen Y, Storrs E, Petralia F, Colaprico A, da Veiga Leprevost F, Pugliese P, Ceccarelli M, Noushmehr H, Nesvizhskii AI, Kamińska B, Priebe W, Lubiński J, Zhang B, Lazar AJ, Kurzawa P, Mesri M, Robles AI, Ding L, Malta TM, Wiznerowicz M. Proteomic-based stemness score measures oncogenic dedifferentiation and enables the identification of druggable targets. CELL GENOMICS 2025:100851. [PMID: 40250426 DOI: 10.1016/j.xgen.2025.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/13/2024] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Cancer progression and therapeutic resistance are closely linked to a stemness phenotype. Here, we introduce a protein-expression-based stemness index (PROTsi) to evaluate oncogenic dedifferentiation in relation to histopathology, molecular features, and clinical outcomes. Utilizing datasets from the Clinical Proteomic Tumor Analysis Consortium across 11 tumor types, we validate PROTsi's effectiveness in accurately quantifying stem-like features. Through integration of PROTsi with multi-omics, including protein post-translational modifications, we identify molecular features associated with stemness and proteins that act as active nodes within transcriptional networks, driving tumor aggressiveness. Proteins highly correlated with stemness were identified as potential drug targets, both shared and tumor specific. These stemness-associated proteins demonstrate predictive value for clinical outcomes, as confirmed by immunohistochemistry in multiple samples. The findings emphasize PROTsi's efficacy as a valuable tool for selecting predictive protein targets, a crucial step in customizing anti-cancer therapy and advancing the clinical development of cures for cancer patients.
Collapse
Affiliation(s)
- Iga Kołodziejczak-Guglas
- International Institute for Molecular Oncology, 60-203 Poznań, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Renan L S Simões
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil
| | - Emerson de Souza Santos
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto 14040-900, Brazil
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital and Laboratory Medicine and Pathobiology, University of Toronto, Toronto ON M5G 1X5, Canada
| | - Rossana N Lazcano Segura
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yifat Geffen
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA 02115, USA
| | - Erik Storrs
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Antonio Colaprico
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Pietro Pugliese
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Houtan Noushmehr
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, MI 48202, USA
| | - Alexey I Nesvizhskii
- Departments of Pathology and Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bożena Kamińska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Waldemar Priebe
- Department of Experimental Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jan Lubiński
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland
| | - Bing Zhang
- Lester and Sue Smith Breast Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paweł Kurzawa
- Department of Oncological Pathology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, 60-514 Poznań, Poland
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD 20850, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD 20850, USA
| | - Li Ding
- Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tathiane M Malta
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto 14040-900, Brazil.
| | - Maciej Wiznerowicz
- International Institute for Molecular Oncology, 60-203 Poznań, Poland; Department of Oncology, Institute of Oncology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, 60-659 Poznań, Poland.
| |
Collapse
|
13
|
Liu J, Guo L, Zhong J, Wu Y, Wang X, Tang X, Min K, Yang Y, Peng W, Wang Q, Ding T, Gu X, Zhang H, Liu Y, Huang C, Cao B, Wang J, Ren L, Yang J. Proteomic Analysis of 442 Clinical Plasma Samples From Individuals With Symptom Records Revealed Subtypes of Convalescent Patients Who Had COVID-19. J Med Virol 2025; 97:e70203. [PMID: 40207927 PMCID: PMC11984345 DOI: 10.1002/jmv.70203] [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: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 04/11/2025]
Abstract
After the coronavirus disease 2019 (COVID-19) pandemic, the postacute effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have gradually attracted attention. To precisely evaluate the health status of convalescent patients with COVID-19, we analyzed symptom and proteome data of 442 plasma samples from healthy controls, hospitalized patients, and convalescent patients 6 or 12 months after SARS-CoV-2 infection. Symptoms analysis revealed distinct relationships in convalescent patients. Results of plasma protein expression levels showed that C1QA, C1QB, C2, CFH, CFHR1, and F10, which regulate the complement system and coagulation, remained highly expressed even at the 12-month follow-up compared with their levels in healthy individuals. By combining symptom and proteome data, 442 plasma samples were categorized into three subtypes: S1 (metabolism-healthy), S2 (COVID-19 retention), and S3 (long COVID). We speculated that convalescent patients reporting hair loss could have a better health status than those experiencing headaches and dyspnea. Compared to other convalescent patients, those reporting sleep disorders, appetite decrease, and muscle weakness may need more attention because they were classified into the S2 subtype, which had the most samples from hospitalized patients with COVID-19. Subtyping convalescent patients with COVID-19 may enable personalized treatments tailored to individual needs. This study provides valuable plasma proteomic datasets for further studies associated with long COVID.
Collapse
Grants
- This work was supported by grants from the National Key R&D Program of China (2023YFC2507102), the Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences, China (CIFMS2022-I2M-1-011, CIFMS2022-I2M-2-001, CIFMS2021-I2M-1-057, CIFMS2021-I2M-1-049, CIFMS2021-I2M-1-044, CIFMS2021-I2M-1-016, CIFMS2021-I2M-1-001, 2022-I2M-CoV19-003, and CIFMS2022-I2M-JB-003), the National Natural Science Foundation of China (82341064), the Haihe Laboratory of Cell Ecosystem Innovation Fund (22HHXBSS00008 and 22HHKYZX0034), and State Key Laboratory Special Fund 2060204.
Collapse
Affiliation(s)
- Jiangfeng Liu
- Haihe Laboratory of Cell EcosystemTianjinChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Li Guo
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
| | - Jingchuan Zhong
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
| | - Yue Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Xinming Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
| | - Xiaoyue Tang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Kaiyuan Min
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Yehong Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Wanjun Peng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Qiaochu Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Tao Ding
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Xiaoying Gu
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Pulmonary and Critical Care MedicineNational Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesBeijingChina
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Pulmonary and Critical Care MedicineCapital Medical UniversityBeijingChina
| | - Hui Zhang
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Pulmonary and Critical Care MedicineNational Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesBeijingChina
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Pulmonary and Critical Care MedicineCapital Medical UniversityBeijingChina
| | - Ying Liu
- Medical DepartmentJin Yin‐Tan HospitalWuhanHubeiChina
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical SciencesWuhanHubeiChina
| | - Chaolin Huang
- Medical DepartmentJin Yin‐Tan HospitalWuhanHubeiChina
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical SciencesWuhanHubeiChina
| | - Bin Cao
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Pulmonary and Critical Care MedicineNational Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesBeijingChina
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Pulmonary and Critical Care MedicineCapital Medical UniversityBeijingChina
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Respiratory Disease PathogenomicsChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lili Ren
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Respiratory Disease PathogenomicsChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Juntao Yang
- Haihe Laboratory of Cell EcosystemTianjinChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
14
|
Cuadrado A, Cazalla E, Bach A, Bathish B, Naidu SD, DeNicola GM, Dinkova-Kostova AT, Fernández-Ginés R, Grochot-Przeczek A, Hayes JD, Kensler TW, León R, Liby KT, López MG, Manda G, Shivakumar AK, Hakomäki H, Moerland JA, Motohashi H, Rojo AI, Sykiotis GP, Taguchi K, Valverde ÁM, Yamamoto M, Levonen AL. Health position paper and redox perspectives - Bench to bedside transition for pharmacological regulation of NRF2 in noncommunicable diseases. Redox Biol 2025; 81:103569. [PMID: 40059038 PMCID: PMC11970334 DOI: 10.1016/j.redox.2025.103569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-activated transcription factor regulating cellular defense against oxidative stress, thereby playing a pivotal role in maintaining cellular homeostasis. Its dysregulation is implicated in the progression of a wide array of human diseases, making NRF2 a compelling target for therapeutic interventions. However, challenges persist in drug discovery and safe targeting of NRF2, as unresolved questions remain especially regarding its context-specific role in diseases and off-target effects. This comprehensive review discusses the dualistic role of NRF2 in disease pathophysiology, covering its protective and/or destructive roles in autoimmune, respiratory, cardiovascular, and metabolic diseases, as well as diseases of the digestive system and cancer. Additionally, we also review the development of drugs that either activate or inhibit NRF2, discuss main barriers in translating NRF2-based therapies from bench to bedside, and consider the ways to monitor NRF2 activation in vivo.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eduardo Cazalla
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Raquel Fernández-Ginés
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28007, Madrid, Spain
| | - Karen T Liby
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Manuela G López
- Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain; Instituto Teófilo Hernando, Madrid, Spain
| | - Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jessica A Moerland
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Graduate School of Medicine Tohoku University, Sendai, Japan; Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Keiko Taguchi
- Laboratory of Food Chemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan; Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Sols-Morreale" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
15
|
Walsh RM, Ambrose J, Jack JL, Eades AE, Bye BA, Tannus Ruckert M, Messaggio F, Olou AA, Chalise P, Pei D, VanSaun MN. Depletion of tumor-derived CXCL5 improves T cell infiltration and anti-PD-1 therapy response in an obese model of pancreatic cancer. J Immunother Cancer 2025; 13:e010057. [PMID: 40121029 PMCID: PMC11931939 DOI: 10.1136/jitc-2024-010057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND CXCR1/2 inhibitors are being implemented with immunotherapies in PDAC clinical trials. CXC-ligands are a family of cytokines responsible for stimulating these receptors; while typically secreted by activated immune cells, fibroblasts, and even adipocytes, they are also secreted by immune-evasive cancer cells. CXC-ligand release is known to occur in response to inflammatory stimuli. Adipose tissue is an endocrine organ and a source of inflammatory signaling peptides. Importantly, adipose-derived cytokines and chemokines are implicated as potential drivers of tumor cell immune evasion; cumulatively, these findings suggest that targeting CXC-ligands may be beneficial in the context of obesity. METHODS RNA-sequencing of human PDAC cell lines was used to assess influences of adipose conditioned media on the cancer cell transcriptome. The adipose-induced secretome of PDAC cells was validated with ELISA for induction of CXCL5 secretion. Human tissue data from CPTAC was used to correlate IL-1β and TNF expression with both CXCL5 mRNA and protein levels. CRISPR-Cas9 was used to knockout CXCL5 from a murine PDAC KPC cell line to assess orthotopic tumor studies in syngeneic, diet-induced obese mice. Flow cytometry and immunohistochemistry were used to compare the immune profiles between tumors with or without CXCL5. Mice-bearing CXCL5 competent or deficient tumors were monitored for differential tumor size in response to anti-PD-1 immune checkpoint blockade therapy. RESULTS Human adipose tissue conditioned media stimulates CXCL5 secretion from PDAC cells via either IL-1β or TNF; neutralization of both is required to significantly block the release of CXCL5 from tumor cells. Ablation of CXCL5 from tumors promoted an enriched immune phenotype with an unanticipatedly increased number of exhausted CD8 T cells. Application of anti-PD-1 treatment to control tumors failed to alter tumor growth, yet treatment of CXCL5-deficient tumors showed response by significantly diminished tumor mass. CONCLUSIONS In summary, our findings show that both TNF and IL-1β can stimulate CXCL5 release from PDAC cells in vitro, which correlates with expression in patient data. CXCL5 depletion in vivo alone is sufficient to promote T cell infiltration into tumors, increasing efficacy and requiring checkpoint blockade inhibition to alleviate tumor burden.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Fanuel Messaggio
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Prabhakar Chalise
- Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
- The University of Kansas Cancer Center, Kansas City, Kansas, USA
| | - Dong Pei
- Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
- The University of Kansas Cancer Center, Kansas City, Kansas, USA
| | - Michael N VanSaun
- Cancer Biology, KUMC, Kansas City, Kansas, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
16
|
Jiang W, Jaehnig EJ, Liao Y, Shi Z, Yaron-Barir TM, Johnson JL, Cantley LC, Zhang B. Deciphering the dark cancer phosphoproteome using machine-learned co-regulation of phosphosites. Nat Commun 2025; 16:2766. [PMID: 40113755 PMCID: PMC11926083 DOI: 10.1038/s41467-025-57993-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Mass spectrometry-based phosphoproteomics offers a comprehensive view of protein phosphorylation, yet our limited knowledge about the regulation and function of most phosphosites hampers the extraction of meaningful biological insights. To address this challenge, we integrate machine learning with phosphoproteomic data from 1195 tumor specimens spanning 11 cancer types to construct CoPheeMap, a network that maps the co-regulation of 26,280 phosphosites. By incorporating network features from CoPheeMap into a second machine learning model, namely CoPheeKSA, we achieve superior performance in predicting kinase-substrate associations. CoPheeKSA uncovers 24,015 associations between 9399 phosphosites and 104 serine/threonine kinases, shedding light on many unannotated phosphosites and understudied kinases. We validate the accuracy of these predictions using experimentally determined kinase-substrate specificities. Through the application of CoPheeMap and CoPheeKSA to phosphosites with high computationally predicted functional significance and those associated with cancer, we demonstrate their effectiveness in systematically elucidating phosphosites of interest. These analyses unveil dysregulated signaling processes in human cancer and identify understudied kinases as potential therapeutic targets.
Collapse
Affiliation(s)
- Wen Jiang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Eric J Jaehnig
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuxing Liao
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zhiao Shi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Tomer M Yaron-Barir
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, 10021, USA
- Englander Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, 10021, USA
- Columbia University Vagelos College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Jared L Johnson
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- Dana Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Lewis C Cantley
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- Dana Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02215, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
17
|
Buttigieg MM, Vlasschaert C, Bick AG, Vanner RJ, Rauh MJ. Inflammatory reprogramming of the solid tumor microenvironment by infiltrating clonal hematopoiesis is associated with adverse outcomes. Cell Rep Med 2025; 6:101989. [PMID: 40037357 PMCID: PMC11970403 DOI: 10.1016/j.xcrm.2025.101989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 12/04/2024] [Accepted: 02/04/2025] [Indexed: 03/06/2025]
Abstract
Clonal hematopoiesis (CH)-the expansion of somatically mutated hematopoietic cells-is common in solid cancers. CH is associated with systemic inflammation, but its impact on tumor biology is underexplored. Here, we report the effects of CH on the tumor microenvironment (TME) using 1,550 treatment-naive patient samples from the Clinical Proteomics Tumor Analysis Consortium (CPTAC) cohort. CH is present in 18.3% of patients, with one-third of CH mutations also detectable in tumor-derived DNA from the same individual (CH-Tum), reflecting CH-mutant leukocyte infiltration. Across cancers, the presence of CH-Tum is associated with worse survival outcomes. Molecular analyses reveal an association between CH-Tum and an immune-rich, inflammatory TME that is notably distinct from age-related gene expression changes. These effects are most prominent in glioblastoma, where CH correlates with pronounced macrophage infiltration, inflammation, and an aggressive, mesenchymal phenotype. Our findings demonstrate that CH shapes the TME, with potential applications as a biomarker in precision oncology.
Collapse
Affiliation(s)
- Marco M Buttigieg
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, Canada
| | | | - Alexander G Bick
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, USA; Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert J Vanner
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Michael J Rauh
- Department of Pathology & Molecular Medicine, Queen's University, Kingston, ON, Canada; Department of Medicine, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
18
|
Schweppe DK, Beliveau BJ, Hoofnagle AN. Molecular Phenotyping With Proteomics. JAMA 2025; 333:898-899. [PMID: 39883454 PMCID: PMC12053805 DOI: 10.1001/jama.2024.28089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
This JAMA Insights explores the capability of proteomics to analyze thousands of proteins in patient samples, which could improve clinicians’ understanding of and ability to treat a wide range of diseases.
Collapse
Affiliation(s)
- Devin K. Schweppe
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | - Brian J. Beliveau
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
- Brotman Baty Institute for Precision Medicine, Seattle, Washington, USA
- Institute of Stem Cell and Regenerative Medicine, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
19
|
Wan X, Zou Y, Zhou Q, Tang Q, Zhu G, Jia L, Yu X, Mo H, Yang X, Wang S. Tumor Prognostic Risk Model Related to Monocytes/Macrophages in Hepatocellular Carcinoma Based on Machine Learning and Multi-Omics. Biol Proced Online 2025; 27:9. [PMID: 40065214 PMCID: PMC11892220 DOI: 10.1186/s12575-025-00270-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Tumor-associated macrophages (TAMs) are crucial in hepatocellular carcinoma (HCC) development and invasion. This study explores monocyte/ macrophage-associated gene expression profiles in HCC, constructs a prognostic model based on these genes, and examines its relationship with drug resistance and immune therapy responses. Single-cell RNA sequencing(scRNA-seq) data from 10 HCC tissue biopsy samples, totaling 24,597 cells, were obtained from the GEO database to identify monocyte/macrophage-associated genes. A prognostic model was constructed and validated using external datasets and Western blot. Relationships between the model, clinical correlates, drug sensitivity, and immune therapy responses were investigated. From scRNA-seq data, 2,799 monocyte/macrophage marker genes were identified. Using the TCGA dataset, a prognostic model based on the single-gene UQCRH was constructed, stratifying patients into high-risk and low-risk groups based on overall survival rates. High-risk group patients showed reduced survival rates and higher UQCRH expression in tumor tissues. Western blot analysis further confirmed the elevated expression of UQCRH in HCC cell lines. Spatial transcriptomics analysis revealed that high UQCRH expression co-localized with malignant cells in the tumor tissue. Drug sensitivity analysis revealed that the high-risk group had lower sensitivity to sorafenib and axitinib. Immune therapy response analysis indicated poorer outcomes in the high-risk group, with more pronounced APC inhibition and a weaker IFN-II response. Clinical indicator analysis showed a positive correlation between high UQCRH expression and tumor invasion. Enrichment analysis of UQCRH and associated molecules indicated involvement in oxidative phosphorylation and mitochondrial electron transport. This study introduces a prognostic model for HCC patients based on monocyte/macrophage marker genes. The single-gene model predicts HCC patient survival and treatment outcomes, identifying high-risk individuals with varying drug sensitivities and immune suppression states.
Collapse
Affiliation(s)
- Xinliang Wan
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Yongchun Zou
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Qichun Zhou
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Qing Tang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Gangxing Zhu
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Luyu Jia
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Xiaoyan Yu
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Handan Mo
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Xiaobing Yang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, 111 Dade Rd, Guangzhou, Guangdong Province, 510120, China.
| | - Sumei Wang
- Clinical and Basic Research Team of TCM Prevention and Treatment of NSCLC, Department of Oncology, The Second Clinical College of Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, 510120, China.
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Chinese Medicine Guangdong Laboratory, 111 Dade Rd, Guangzhou, Guangdong Province, 510120, China.
| |
Collapse
|
20
|
Fang M, Wu Z, Xia Z, Xiao J. Diagnostic, prognostic, and immunological roles of NCAPG in pan-cancer: A bioinformatics analysis. Medicine (Baltimore) 2025; 104:e41761. [PMID: 40068055 PMCID: PMC11903004 DOI: 10.1097/md.0000000000041761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 11/13/2024] [Accepted: 02/16/2025] [Indexed: 03/14/2025] Open
Abstract
Growing studies have shown that non-SMC condensin I complex subunit G (NCAPG) was highly expressed in a variety of tumors and was involved in the progression of multitumors, but the role of NCAPG in tumorigenesis is not fully understood. Our study purposed to systematically investigate the role of NCAPG across cancer types. Interacting molecules with NCAPG were analyzed using searching bioinformatics websites including Search Tool for the Retrieval of Interacting Genes/Proteins, GeneMANIA, and Global Positioning System-Prot. NCAPG-related diseases were acquired using the Open Targets Platform. The interaction of NCAPG and 14 cancer functional states was achieved using the CancerSEA website. The databases including the University of California Santa Cruz Xena, Genotype-Tissue Expression, The Cancer Genome Atlas Program, Human Protein Atlas, and XIANTAO Academic were used to interpret the expression of NCAPG. Correlations between NCAPG expression and immune infiltration and immune-related molecules were analyzed by using Tumor Immune Estimation Resource Version 2 and Tumor and Immune System Interaction Database databases. NCAPG expression was significantly upregulated in most cancer types. NCAPG was identified as a marker of diagnostic value and prognostic significance in most cancer types. NCAPG expression was related to immune cell infiltration and immune-related molecules across various cancers, especially kidney renal clear cell carcinoma and thyroid carcinoma. Furthermore, NCAPG expression could affect the enrichment and decrease immune cell infiltration to influence prognosis in kidney renal clear cell carcinoma but was devoid of evidence in thyroid carcinoma. NCAPG was a prospective marker for the diagnosis and prognosis of pan-cancer. Our results suggested that NCAPG was a potential cancer biomarker for the diagnosis and prognosis of pan-cancer. NCAPG might affect the immune microenvironment, which could be applied in the development of new-targeted drugs for immunotherapy.
Collapse
Affiliation(s)
- Min Fang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
- “The 14th Five-Year Plan” Application Characteristic Discipline of Hunan Province (Pharmaceutical Science) Changsha Medical University, Changsha, China
| | - Zhu Wu
- Hunan Provincial Key Laboratory of the Traditional Chinese Medicine Agricultural Biogenomics, Changsha Medical University, Changsha, China
| | - Zhi Xia
- Department of Oncology, Hunan Provincial People’s Hospital, First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Jian Xiao
- Department of Geriatric Respiratory and Critical Care Medicine, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
21
|
Zheng X, Mund A, Mann M. Deciphering functional tumor-immune crosstalk through highly multiplexed imaging and deep visual proteomics. Mol Cell 2025; 85:1008-1023.e7. [PMID: 39814024 DOI: 10.1016/j.molcel.2024.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/05/2024] [Accepted: 12/20/2024] [Indexed: 01/18/2025]
Abstract
Deciphering the intricate tumor-immune interactions within the microenvironment is crucial for advancing cancer immunotherapy. Here, we introduce mipDVP, an advanced approach integrating highly multiplexed imaging, single-cell laser microdissection, and sensitive mass spectrometry to spatially profile the proteomes of distinct cell populations in a human colorectal and tonsil cancer with high sensitivity. In a colorectal tumor-a representative cold tumor-we uncovered spatial compartmentalization of an immunosuppressive macrophage barrier that potentially impedes T cell infiltration. Spatial proteomic analysis revealed distinct functional states of T cells in different tumor compartments. In a tonsil cancer sample-a hot tumor-we identified significant proteomic heterogeneity among cells influenced by proximity to cytotoxic T cell subtypes. T cells in the tumor parenchyma exhibit metabolic adaptations to hypoxic regions. Our spatially resolved, highly multiplexed strategy deciphers the complex cellular interplay within the tumor microenvironment, offering valuable insights for identifying immunotherapy targets and predictive signatures.
Collapse
Affiliation(s)
- Xiang Zheng
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen 2200, Copenhagen, Denmark; Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark.
| | - Andreas Mund
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen 2200, Copenhagen, Denmark; OmicVision Biosciences, BioInnovation Institute, Copenhagen 2200, Denmark
| | - Matthias Mann
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen 2200, Copenhagen, Denmark; Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany.
| |
Collapse
|
22
|
Cheng H, Liang Z, Wu Y, Hu J, Cao B, Liu Z, Liu B, Cheng H, Liu ZX. Inferring kinase-phosphosite regulation from phosphoproteome-enriched cancer multi-omics datasets. Brief Bioinform 2025; 26:bbaf143. [PMID: 40194556 PMCID: PMC11975364 DOI: 10.1093/bib/bbaf143] [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: 11/07/2024] [Revised: 02/03/2025] [Accepted: 03/14/2025] [Indexed: 04/09/2025] Open
Abstract
Phosphorylation in eukaryotic cells plays a key role in regulating cell signaling and disease progression. Despite the ability to detect thousands of phosphosites in a single experiment using high-throughput technologies, the kinases responsible for regulating these sites are largely unidentified. To solve this, we collected the quantitative data at the transcriptional, protein, and phosphorylation levels of 10 159 samples from 23 tumor datasets and 15 adjacent normal tissue datasets. Our analysis aimed to uncover the potential impact and linkage of kinase-phosphosite (KPS) pairs through experimental evidence in publications and prediction tools commonly used. We discovered that both experimentally validated and tool-predicted KPS pairs were enriched in groups where there is a significant correlation between kinase expression/phosphorylation level and the phosphorylation level of phosphosite. This suggested that a quantitative correlation could infer the KPS interconnections. Furthermore, the Spearman's correlation coefficient for these pairs were notably higher in tumor samples, indicating that these regulatory interactions are particularly pronounced in tumors. Consequently, building on the KPS correlations of different datasets as predictive features, we have developed an innovative approach that employed an oversampling method combined with and XGBoost algorithm (SMOTE-XGBoost) to predict potential kinase-specific phosphorylation sites in proteins. Moreover, the computed correlations and predictions of kinase-phosphosite interconnections were integrated into the eKPI database (https://ekpi.omicsbio.info/). In summary, our study could provide helpful information and facilitate further research on the regulatory relationship between kinases and phosphosites.
Collapse
Affiliation(s)
- Haoyang Cheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
- Department of Computer Science, The University of Hong Kong, Pokfulam Road, Hong Kong Special Administrative Region 999077, China
| | - Zhuoran Liang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yijin Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Jiamin Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Bijin Cao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
- School of Life Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Zekun Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Bo Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
- School of Life Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Han Cheng
- School of Life Sciences, Zhengzhou University, 100 Science Avenue, Zhengzhou 450001, China
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| |
Collapse
|
23
|
Killarney ST, Mesa G, Washart R, Mayro B, Dillon K, Wardell SE, Newlin M, Lu M, Rmaileh AA, Liu N, McDonnell DP, Pendergast AM, Wood KC. PKN2 Is a Dependency of the Mesenchymal-like Cancer Cell State. Cancer Discov 2025; 15:595-615. [PMID: 39560431 PMCID: PMC11875962 DOI: 10.1158/2159-8290.cd-24-0928] [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: 06/27/2024] [Revised: 10/11/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
Cancer cells exploit a mesenchymal-like transcriptional state (MLS) to survive drug treatments. Although the MLS is well characterized, few therapeutic vulnerabilities targeting this program have been identified. In this study, we systematically identify the dependency network of mesenchymal-like cancers through an analysis of gene essentiality scores in ∼800 cancer cell lines, nominating a poorly studied kinase, PKN2, as a top therapeutic target of the MLS. Coessentiality relationships, biochemical experiments, and genomic analyses of patient tumors revealed that PKN2 promotes mesenchymal-like cancer growth through a PKN2-SAV1-TAZ signaling mechanism. Notably, pairing genetic PKN2 inhibition with clinically relevant targeted therapies against EGFR, KRAS, and BRAF suppresses drug resistance by depleting mesenchymal-like drug-tolerant persister cells. These findings provide evidence that PKN2 is a core regulator of the Hippo tumor suppressor pathway and highlight the potential of PKN2 inhibition as a generalizable therapeutic strategy to overcome drug resistance driven by the MLS across cancer contexts. Significance: This work identifies PKN2 as a core member of the Hippo signaling pathway, and its inhibition blocks YAP/TAZ-driven tumorigenesis. Furthermore, this study discovers PKN2-TAZ as arguably the most selective dependency of mesenchymal-like cancers and supports specific inhibition of PKN2 as a provocative strategy to overcome drug resistance in diverse cancer contexts. See related commentary by Shen and Tan, p. 458.
Collapse
Affiliation(s)
- Shane T. Killarney
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Gabriel Mesa
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Rachel Washart
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Benjamin Mayro
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kerry Dillon
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Suzanne E. Wardell
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Madeline Newlin
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Min Lu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Areej Abu Rmaileh
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Nicky Liu
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | | | | | - Kris C. Wood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| |
Collapse
|
24
|
Hu GS, Zheng ZZ, He YH, Wang DC, Nie RC, Liu W. Integrated Analysis of Proteome and Transcriptome Profiling Reveals Pan-Cancer-Associated Pathways and Molecular Biomarkers. Mol Cell Proteomics 2025; 24:100919. [PMID: 39884577 PMCID: PMC11907456 DOI: 10.1016/j.mcpro.2025.100919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
Understanding dysregulated genes and pathways in cancer is critical for precision oncology. Integrating mass spectrometry-based proteomic data with transcriptomic data presents unique opportunities for systematic analyses of dysregulated genes and pathways in pan-cancer. Here, we compiled a comprehensive set of datasets, encompassing proteomic data from 2404 samples and transcriptomic data from 7752 samples across 13 cancer types. Comparisons between normal or adjacent normal tissues and tumor tissues identified several dysregulated pathways including mRNA splicing, interferon pathway, fatty acid metabolism, and complement coagulation cascade in pan-cancer. Additionally, pan-cancer upregulated and downregulated genes (PCUGs and PCDGs) were also identified. Notably, RRM2 and ADH1B, two genes which belong to PCUGs and PCDGs, respectively, were identified as robust pan-cancer diagnostic biomarkers. TNM stage-based comparisons revealed dysregulated genes and biological pathways involved in cancer progression, among which the dysregulation of complement coagulation cascade and epithelial-mesenchymal transition are frequent in multiple types of cancers. A group of pan-cancer continuously upregulated and downregulated proteins in different tumor stages (PCCUPs and PCCDPs) were identified. We further constructed prognostic risk stratification models for corresponding cancer types based on dysregulated genes, which effectively predict the prognosis for patients with these cancers. Drug prediction based on PCUGs and PCDGs as well as PCCUPs and PCCDPs revealed that small molecule inhibitors targeting CDK, HDAC, MEK, JAK, PI3K, and others might be effective treatments for pan-cancer, thereby supporting drug repurposing. We also developed web tools for cancer diagnosis, pathologic stage assessment, and risk evaluation. Overall, this study highlights the power of combining proteomic and transcriptomic data to identify valuable diagnostic and prognostic markers as well as drug targets and treatments for cancer.
Collapse
Affiliation(s)
- Guo-Sheng Hu
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, China; State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zao-Zao Zheng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yao-Hui He
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Du-Chuang Wang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Rui-Chao Nie
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wen Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
25
|
Huang M, Cai J, Yang S, Zhao Q, Shao Z, Zhang F, Zhang Y, Cao A, Li D. Secernin-2 Stabilizes Histone Methyltransferase KMT2C to Suppress Progression and Confer Therapeutic Sensitivity to PARP Inhibition in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413280. [PMID: 39836524 PMCID: PMC11905051 DOI: 10.1002/advs.202413280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/24/2024] [Indexed: 01/23/2025]
Abstract
Triple-negative breast cancer (TNBC) is a difficulty and bottleneck in the clinical treatment of breast cancer due to a lack of effective therapeutic targets. Herein, we first report that secernin 2 (SCRN2), an uncharacterized gene in human cancer, acts as a novel tumor suppressor in TNBC to inhibit cancer progression and enhance therapeutic sensitivity to poly(ADP-ribose) polymerase (PARP) inhibition both in vitro and in vivo. SCRN2 is downregulated in TNBC through chaperone-mediated autophagic degradation, and its downregulation is associated with poor patient prognosis. Moreover, SCRN2 impedes the proteasomal degradation of histone-lysine N-methyltransferase 2C (KMT2C) by recruiting Bcl2-associated athanogene 2 to block the interaction of KMT2C with E3 ubiquitin-protein ligase CHIP. Consistently, SCRN2 transcriptionally activates Bcl2-modifying factor by amplifying histone H3 monomethylation at lysine 4 at its enhancer, thereby inducing intrinsic apoptosis. Notably, KMT2C knockdown restores the impaired TNBC progression caused by SCRN2 overexpression both in vitro and in vivo. Furthermore, SCRN2 decreases the expression of key DNA repair-related genes and induces endogenous DNA damage, thus conferring therapeutic sensitivity of TNBC cells to PARP inhibition. Collectively, these findings identify SCRN2 as a novel suppressor of TNBC, reveal its mechanism of action, and highlight its potential role in TNBC therapy.
Collapse
Affiliation(s)
- Min‐Ying Huang
- Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Cancer Institute, Shanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jia‐Yang Cai
- Department of Breast SurgeryShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Shao‐Ying Yang
- Cancer Institute, Shanghai Medical CollegeFudan UniversityShanghai200032China
| | - Qian Zhao
- Cancer Institute, Shanghai Medical CollegeFudan UniversityShanghai200032China
- Department of Breast SurgeryShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Zhi‐Min Shao
- Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Cancer Institute, Shanghai Medical CollegeFudan UniversityShanghai200032China
- Department of Breast SurgeryShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Key Laboratory of Breast CancerShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Key Laboratory of Radiation OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Fang‐Lin Zhang
- Cancer Institute, Shanghai Medical CollegeFudan UniversityShanghai200032China
| | - Yin‐Ling Zhang
- Cancer Institute, Shanghai Medical CollegeFudan UniversityShanghai200032China
| | - A‐Yong Cao
- Department of Breast SurgeryShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Da‐Qiang Li
- Shanghai Cancer Center and Institutes of Biomedical SciencesShanghai Medical CollegeFudan UniversityShanghai200032China
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
- Cancer Institute, Shanghai Medical CollegeFudan UniversityShanghai200032China
- Department of Breast SurgeryShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Key Laboratory of Breast CancerShanghai Medical CollegeFudan UniversityShanghai200032China
- Shanghai Key Laboratory of Radiation OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| |
Collapse
|
26
|
Bökenkamp JE, Keuper K, Redel S, Barthel K, Johnson L, Becker A, Wieland A, Räschle M, Storchová Z. Proteogenomic analysis reveals adaptive strategies for alleviating the consequences of aneuploidy in cancer. EMBO J 2025; 44:1829-1865. [PMID: 39930267 PMCID: PMC11914506 DOI: 10.1038/s44318-025-00372-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 03/19/2025] Open
Abstract
Aneuploidy is prevalent in cancer and associates with fitness advantage and poor patient prognosis. Yet, experimentally induced aneuploidy initially leads to adverse effects and impaired proliferation, suggesting that cancer cells must adapt to aneuploidy. We performed in vitro evolution of cells with extra chromosomes and obtained cell lines with improved proliferation and gene expression changes congruent with changes in aneuploid cancers. Integrated analysis of cancer multi-omics data and model cells revealed increased expression of DNA replicative and repair factors, reduced genomic instability, and reduced lysosomal degradation. We identified E2F4 and FOXM1 as transcription factors strongly associated with adaptation to aneuploidy in vitro and in cancers and validated this finding. The adaptation to aneuploidy also coincided with specific copy number aberrations that correlate with poor patient prognosis. Chromosomal engineering mimicking these aberrations improved aneuploid cell proliferation, while loss of previously present extra chromosomes impaired it. The identified common adaptation strategies suggest replication stress, genomic instability, and lysosomal stress as common liabilities of aneuploid cancers.
Collapse
Affiliation(s)
- Jan-Eric Bökenkamp
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Kristina Keuper
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
- Danish Cancer Institute, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Stefan Redel
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Karen Barthel
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Leah Johnson
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Amelie Becker
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Angela Wieland
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Markus Räschle
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany
| | - Zuzana Storchová
- RPTU Kaiserslautern-Landau, Paul- Ehrlich Strasse 24, 67663, Kaiserslautern, Germany.
| |
Collapse
|
27
|
Suo Y, Song Y, Wang Y, Liu Q, Rodriguez H, Zhou H. Advancements in proteogenomics for preclinical targeted cancer therapy research. BIOPHYSICS REPORTS 2025; 11:56-76. [PMID: 40070661 PMCID: PMC11891078 DOI: 10.52601/bpr.2024.240053] [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: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 03/14/2025] Open
Abstract
Advancements in molecular characterization technologies have accelerated targeted cancer therapy research at unprecedented resolution and dimensionality. Integrating comprehensive multi-omic molecular profiling of a tumor, proteogenomics, marks a transformative milestone for preclinical cancer research. In this paper, we initially provided an overview of proteogenomics in cancer research, spanning genomics, transcriptomics, and proteomics. Subsequently, the applications were introduced and examined from different perspectives, including but not limited to genetic alterations, molecular quantifications, single-cell patterns, different post-translational modification levels, subtype signatures, and immune landscape. We also paid attention to the combined multi-omics data analysis and pan-cancer analysis. This paper highlights the crucial role of proteogenomics in preclinical targeted cancer therapy research, including but not limited to elucidating the mechanisms of tumorigenesis, discovering effective therapeutic targets and promising biomarkers, and developing subtype-specific therapies.
Collapse
Affiliation(s)
- Yuying Suo
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanli Song
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuqiu Wang
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Qian Liu
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, National Institutes of Health, Rockville, MD 20850, USA
| | - Hu Zhou
- Department of Analytical Chemistry, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| |
Collapse
|
28
|
Chen Y, Ali M, Tayyab MB, Nazir MM, Umar M, Khan S, Ismail DM, Abdel-Maksoud MA, Ebaid H, Alamri A, Almutairi S, Almanaa TN, Kiani BH. The role of Prolyl 3-Hydroxylase 1 (P3H1) in tumor development and prognosis: a pan-cancer analysis with validation in colonic adenocarcinoma. Am J Transl Res 2025; 17:770-790. [PMID: 40092085 PMCID: PMC11909568 DOI: 10.62347/suvc1606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 12/16/2024] [Indexed: 03/19/2025]
Abstract
BACKGROUND Cancer is a multifaceted disease characterized by unregulated cell proliferation, evasion of apoptosis, and metastasis. Recent studies have highlighted the importance of extracellular matrix remodeling and post-translational modifications in tumorigenesis. Prolyl 3-hydroxylase 1 (P3H1), an enzyme involved in collagen hydroxylation, has gained attention for its role in cancer progression. METHODS This study investigates P3H1 expression, prognostic value, and functional relevance across multiple human cancers using a combination of bioinformatic and experimental approaches. RESULTS Using The Cancer Genome Atlas (TCGA) data from TIMER2.0 and UALCAN databases, we observed a significant upregulation of P3H1 mRNA and protein in various cancers. Prognostic analysis using GEPIA2 and KM plotter revealed that high P3H1 expression correlates with poorer overall survival in colon adenocarcinoma (COAD), kidney renal clear cell carcinoma (KIRC), and liver hepatocellular carcinoma (LIHC). Further, genetic and promoter methylation analyses showed low mutation frequencies and reduced methylation of P3H1 in specific cancer types. Functional and pathway enrichment analyses indicated that P3H1 is involved in collagen formation, endoplasmic reticulum activity, and pathways such as ECM-receptor interaction and PI3K-Akt signaling. Validation by enzyme linked immunosorbent assay in COAD patient serum samples demonstrated significantly elevated P3H1 levels compared to healthy controls, with an AUC approaching 1.0 by receiver operating characteristic (ROC) curve analysis. This suggests its potential as a diagnostic biomarker. Additionally, functional experiments were conducted in COAD cells to assess P3H1's role in tumorigenesis. Knockdown of P3H1 in HCT116 cells resulted in a significant reduction in cell proliferation, colony formation, and migratory abilities of these cells. CONCLUSION These findings emphasize P3H1's relevance in COAD, KIRC, and LIHC pathogenesis and possible utility in clinical diagnosis.
Collapse
Affiliation(s)
- Yanqin Chen
- Nanjing Drum Tower HospitalNanjing 210000, Jiangsu, China
| | - Moazzam Ali
- Department of Gastroenterology, Hayatabad Medical Complex PeshawarPeshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Bilal Tayyab
- Institute of Drug Discovery and Development, Zhengzhou UniversityZhengzhou 450001, Henan, China
| | | | - Muhammad Umar
- Department of Neurosurgery, Allied Hospital FaisalabadFaisalabad 37521, Punjab, Pakistan
| | - Salman Khan
- DHQ Teaching Hospital, GMCDikah, Abbottabad 22010, Khyber Pakhtunkhwa, Pakistan
| | | | - Mostafa A Abdel-Maksoud
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh 11451, Saudi Arabia
| | - Hossam Ebaid
- Department of Zoology, College of Science, King Saud UniversityP.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud UniversityP.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Saeedah Almutairi
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh 11451, Saudi Arabia
| | - Taghreed N Almanaa
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh 11451, Saudi Arabia
| | - Bushra Hafeez Kiani
- Department of Biology and Biotechnology, Worcester Polytechnic InstituteWorcester, Massachuesetts 01609, USA
| |
Collapse
|
29
|
Paton V, Türei D, Ivanova O, Müller-Dott S, Rodriguez-Mier P, Venafra V, Perfetto L, Garrido-Rodriguez M, Saez-Rodriguez J. NetworkCommons: bridging data, knowledge, and methods to build and evaluate context-specific biological networks. Bioinformatics 2025; 41:btaf048. [PMID: 39907203 PMCID: PMC11846666 DOI: 10.1093/bioinformatics/btaf048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/22/2025] [Accepted: 01/31/2025] [Indexed: 02/06/2025] Open
Abstract
SUMMARY We present NetworkCommons, a platform for integrating prior knowledge, omics data, and network inference methods, facilitating their usage and evaluation. NetworkCommons aims to be an infrastructure for the network biology community that supports the development of better methods and benchmarks, by enhancing interoperability and integration. AVAILABILITY AND IMPLEMENTATION NetworkCommons is implemented in Python and offers programmatic access to multiple omics datasets, network inference methods, and benchmarking setups. It is a free software, available at https://github.com/saezlab/networkcommons, and deposited in Zenodo at https://doi.org/10.5281/zenodo.14719118.
Collapse
Affiliation(s)
- Victor Paton
- Institute for Computational Biomedicine, Heidelberg University Hospital and Heidelberg University, Heidelberg, 69120, Germany
| | - Denes Türei
- Institute for Computational Biomedicine, Heidelberg University Hospital and Heidelberg University, Heidelberg, 69120, Germany
| | - Olga Ivanova
- Institute for Computational Biomedicine, Heidelberg University Hospital and Heidelberg University, Heidelberg, 69120, Germany
| | - Sophia Müller-Dott
- Institute for Computational Biomedicine, Heidelberg University Hospital and Heidelberg University, Heidelberg, 69120, Germany
| | - Pablo Rodriguez-Mier
- Institute for Computational Biomedicine, Heidelberg University Hospital and Heidelberg University, Heidelberg, 69120, Germany
| | - Veronica Venafra
- Department of Biology and Biotechnologies “C. Darwin”, University of Rome La Sapienza, 00185 Rome, Italy
- Ph.D. Program in Cellular and Molecular Biology, Department of Biology, University of Rome ‘Tor Vergata’, 00133 Rome, Italy
| | - Livia Perfetto
- Department of Biology and Biotechnologies “C. Darwin”, University of Rome La Sapienza, 00185 Rome, Italy
| | - Martin Garrido-Rodriguez
- Institute for Computational Biomedicine, Heidelberg University Hospital and Heidelberg University, Heidelberg, 69120, Germany
- Molecular Systems Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, 69117, Germany
- European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridgeshire, CB10 1SD, United Kingdom
| | - Julio Saez-Rodriguez
- Institute for Computational Biomedicine, Heidelberg University Hospital and Heidelberg University, Heidelberg, 69120, Germany
- European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridgeshire, CB10 1SD, United Kingdom
| |
Collapse
|
30
|
Guo T, Steen JA, Mann M. Mass-spectrometry-based proteomics: from single cells to clinical applications. Nature 2025; 638:901-911. [PMID: 40011722 DOI: 10.1038/s41586-025-08584-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 01/02/2025] [Indexed: 02/28/2025]
Abstract
Mass-spectrometry (MS)-based proteomics has evolved into a powerful tool for comprehensively analysing biological systems. Recent technological advances have markedly increased sensitivity, enabling single-cell proteomics and spatial profiling of tissues. Simultaneously, improvements in throughput and robustness are facilitating clinical applications. In this Review, we present the latest developments in proteomics technology, including novel sample-preparation methods, advanced instrumentation and innovative data-acquisition strategies. We explore how these advances drive progress in key areas such as protein-protein interactions, post-translational modifications and structural proteomics. Integrating artificial intelligence into the proteomics workflow accelerates data analysis and biological interpretation. We discuss the application of proteomics to single-cell analysis and spatial profiling, which can provide unprecedented insights into cellular heterogeneity and tissue architecture. Finally, we examine the transition of proteomics from basic research to clinical practice, including biomarker discovery in body fluids and the promise and challenges of implementing proteomics-based diagnostics. This Review provides a broad and high-level overview of the current state of proteomics and its potential to revolutionize our understanding of biology and transform medical practice.
Collapse
Affiliation(s)
- Tiannan Guo
- State Key Laboratory of Medical Proteomics, School of Medicine, Westlake University, Hangzhou, China.
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Research Center for Industries of the Future, School of Life Sciences, Westlake University, Hangzhou, China.
| | - Judith A Steen
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, USA.
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
- NNF Center for Protein Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
31
|
Parry TL, Gilmore LA, Khamoui AV. Pan-cancer secreted proteome and skeletal muscle regulation: insight from a proteogenomic data-driven knowledge base. Funct Integr Genomics 2025; 25:14. [PMID: 39812750 DOI: 10.1007/s10142-024-01524-7] [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: 09/20/2024] [Revised: 12/16/2024] [Accepted: 12/31/2024] [Indexed: 01/16/2025]
Abstract
Large-scale, pan-cancer analysis is enabled by data driven knowledge bases that link tumor molecular profiles with phenotypes. A debilitating cancer-related phenotype is skeletal muscle loss, or cachexia, which occurs partly from tumor products secreted into circulation. Using the LinkedOmicsKB knowledge base assembled from the Clinical Proteomics Tumor Analysis Consortium proteogenomic analysis, along with catalogs of human secretome proteins, ligand-receptor pairs and molecular signatures, we sought to identify candidate pan-cancer proteins secreted to blood that could regulate skeletal muscle phenotypes in multiple solid cancers. Tumor proteins having significant pan-cancer associations with muscle were referenced against secretome proteins secreted to blood from the Human Protein Atlas, then verified as increased in paired tumor vs. normal tissues in pan-cancer manner. This workflow revealed seven secreted proteins from cancers afflicting kidneys, head and neck, lungs and pancreas that classified as protein-binding activity modulator, extracellular matrix protein or intercellular signaling molecule. Concordance of these biomarkers with validated molecular signatures of cachexia and senescence supported relevance to muscle and cachexia disease biology, and high tumor expression of the biomarker set associated with lower overall survival. In this article, we discuss avenues by which skeletal muscle and cachexia may be regulated by these candidate pan-cancer proteins secreted to blood, and conceptualize a strategy that considers them collectively as a biomarker signature with potential for refinement by data analytics and radiogenomics for predictive testing of future risk in a non-invasive, blood-based panel amenable to broad uptake and early management.
Collapse
Affiliation(s)
- Traci L Parry
- Department of Kinesiology, University of North Carolina Greensboro, Greensboro, NC, USA
| | - L Anne Gilmore
- Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Human Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andy V Khamoui
- Department of Exercise Science and Health Promotion, Florida Atlantic University, Boca Raton, FL, USA.
- Institute for Human Health and Disease Intervention, Florida Atlantic University, Jupiter, FL, USA.
- Stiles-Nicholson Brain Institute, Florida Atlantic University, Jupiter, FL, USA.
| |
Collapse
|
32
|
Rawal O, Turhan B, Peradejordi IF, Chandrasekar S, Kalayci S, Gnjatic S, Johnson J, Bouhaddou M, Gümüş ZH. PhosNetVis: A web-based tool for fast kinase-substrate enrichment analysis and interactive 2D/3D network visualizations of phosphoproteomics data. PATTERNS (NEW YORK, N.Y.) 2025; 6:101148. [PMID: 39896259 PMCID: PMC11783894 DOI: 10.1016/j.patter.2024.101148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/12/2024] [Accepted: 12/11/2024] [Indexed: 02/04/2025]
Abstract
Protein phosphorylation involves the reversible modification of a protein (substrate) residue by another protein (kinase). Liquid chromatography-mass spectrometry studies are rapidly generating massive protein phosphorylation datasets across multiple conditions. Researchers then must infer kinases responsible for changes in phosphosites of each substrate. However, tools that infer kinase-substrate interactions (KSIs) are not optimized to interactively explore the resulting large and complex networks, significant phosphosites, and states. There is thus an unmet need for a tool that facilitates user-friendly analysis, interactive exploration, visualization, and communication of phosphoproteomics datasets. We present PhosNetVis, a web-based tool for researchers of all computational skill levels to easily infer, generate, and interactively explore KSI networks in 2D or 3D by streamlining phosphoproteomics data analysis steps within a single tool. PhostNetVis lowers barriers for researchers by rapidly generating high-quality visualizations to gain biological insights from their phosphoproteomics datasets. It is available at https://gumuslab.github.io/PhosNetVis/.
Collapse
Affiliation(s)
- Osho Rawal
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Berk Turhan
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Faculty of Engineering and Natural Sciences, Sabanci University, 34956 Istanbul, Türkiye
| | - Irene Font Peradejordi
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cornell Tech, Cornell University, New York, NY 10044, USA
| | - Shreya Chandrasekar
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cornell Tech, Cornell University, New York, NY 10044, USA
| | - Selim Kalayci
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sacha Gnjatic
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeffrey Johnson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zeynep H. Gümüş
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
33
|
Yang ZZ, Yang B, Yan H, Ma X, Tian B, Zheng B, Chen YX, Dong YM, Deng J, Zhan Z, Shi Y, Zhang JY, Lu D, He JH, Zhang Y, Hu K, Zhu S, Zhou K, Zhang YC, Zheng Y, Yin D, Liao JY. DCAF13-mediated K63-linked ubiquitination of RNA polymerase I promotes uncontrolled proliferation in Breast Cancer. Nat Commun 2025; 16:557. [PMID: 39788980 PMCID: PMC11718263 DOI: 10.1038/s41467-025-55851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 12/31/2024] [Indexed: 01/12/2025] Open
Abstract
Hyperactivation of ribosome biogenesis (RiBi) drives cancer progression, yet the role of RiBi-associated proteins (RiBPs) in breast cancer (BC) is underexplored. In this study, we perform a comprehensive multi-omics analysis and reveal that assembly and maturation factors (AMFs), a subclass of RiBPs, are upregulated at both RNA and protein levels in BC, correlating with poor patient outcomes. In contrast, ribosomal proteins (RPs) do not show systematic upregulation across various cancers, including BC. We further demonstrate that the oncogenic activation of a top AMF candidate in BC, DCAF13, enhances Pol I transcription and promotes proliferation in BC cells both in vitro and in vivo. Mechanistically, DCAF13 promotes Pol I transcription activity by facilitating the K63-linked ubiquitination of RPA194. This process stimulates global protein synthesis and cell growth. Our findings uncover a modification of RPA194 that regulates Pol I activity; this modification is dysregulated in BC, contributing to cancer progression.
Collapse
Affiliation(s)
- Zhi-Zhi Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Bing Yang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Haiyan Yan
- Department of Clinical Laboratory, Shenshan Central Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, 516600, PR China
| | - Xingyu Ma
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Bin Tian
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Bingqi Zheng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Yong-Xian Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Yi-Ming Dong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Jinsi Deng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Ziling Zhan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Yanmei Shi
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Jing Yuan Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Daning Lu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Jie-Hua He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Yin Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - KaiShun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Shuang Zhu
- Center for Bioresources and Drug Discovery and School of Biosciences and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Keda Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, PR China
| | - Yu-Chan Zhang
- Guangdong Provincial Key Laboratory of Plant Resources, State Key Laboratory for Biocontrol, School of Life Science, Sun Yat-Sen University, Guangzhou, 510275, PR China
| | - Yiqing Zheng
- Center for Precision Medicine, Shenshan Central Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, 516600, PR China.
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107 West Yan Jiang Road, Guangzhou, 510120, PR China.
- Institute of Hearing and Speech-Language Science, Sun Yat-sen University, 107 West Yan Jiang Road, Guangzhou, 510120, PR China.
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China.
| | - Jian-You Liao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China.
- Center for Precision Medicine, Shenshan Central Hospital, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong, 516600, PR China.
| |
Collapse
|
34
|
Ma L, Li Y, Wu J, Gao Y. Bioinformatics approaches to multi-omics analysis of the potential of CDKN2A as a biomarker and therapeutic target for uterine corpus endometrial carcinoma. Sci Rep 2025; 15:895. [PMID: 39762354 PMCID: PMC11704072 DOI: 10.1038/s41598-025-85364-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Uterine corpus endometrial carcinoma (UCEC) is a significant cause of cancer-related mortality among women worldwide. Prior research has demonstrated an association between cyclin-dependent kinase inhibitor 2 A (CDKN2A) and various tumors. As a member of the INK4 family, CDKN2A is involved in cell cycle regulation by controlling CDKs. In the present study, bioinformatics was used to analyze public datasets. The expression levels, signaling pathways, and copy number variations of CDKN2A in UCEC were explored, along with its immune cell subset associations. CDKN2A expression was found to be elevated in UCEC, particularly in the signaling pathways involved in cell proliferation and inflammation. Analysis of somatic copy number alterations in the TCGA (The Cancer Genome Atlas)-UCEC dataset revealed a connection between CDKN2A and drug metabolism in UCEC. Assessment of the relationship between CDKN2A and genes involved in immunotherapy for UCEC patients showed a negative correlation between CDKN2A and CD8+ T cell activity, as well as IL-2 and TP53. Collectively, these insights suggest that CDKN2A may be a potential biomarker for prognosis and treatment strategies in UCEC.
Collapse
Affiliation(s)
- Liang Ma
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400000, China
| | - Yuling Li
- Biochemistry and Molecular Biology, College of Basic Medical Science, Chongqing Medical University, Chongqing, 400000, China
| | - Jingxian Wu
- Department of Pathology, College of Basic Medicine, Chongqing Medical University, Chongqing, 400000, China.
- Molecular Medicine Diagnostic and Testing Center, Chongqing Medical University, Chongqing, 400000, China.
- Department of Pathology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China.
| | - Yanfei Gao
- Biochemistry and Molecular Biology, College of Basic Medical Science, Chongqing Medical University, Chongqing, 400000, China.
| |
Collapse
|
35
|
Shi Z, Lei JT, Elizarraras JM, Zhang B. Mapping the functional network of human cancer through machine learning and pan-cancer proteogenomics. NATURE CANCER 2025; 6:205-222. [PMID: 39663389 PMCID: PMC12036749 DOI: 10.1038/s43018-024-00869-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 10/25/2024] [Indexed: 12/13/2024]
Abstract
Large-scale omics profiling has uncovered a vast array of somatic mutations and cancer-associated proteins, posing substantial challenges for their functional interpretation. Here we present a network-based approach centered on FunMap, a pan-cancer functional network constructed using supervised machine learning on extensive proteomics and RNA sequencing data from 1,194 individuals spanning 11 cancer types. Comprising 10,525 protein-coding genes, FunMap connects functionally associated genes with unprecedented precision, surpassing traditional protein-protein interaction maps. Network analysis identifies functional protein modules, reveals a hierarchical structure linked to cancer hallmarks and clinical phenotypes, provides deeper insights into established cancer drivers and predicts functions for understudied cancer-associated proteins. Additionally, applying graph-neural-network-based deep learning to FunMap uncovers drivers with low mutation frequency. This study establishes FunMap as a powerful and unbiased tool for interpreting somatic mutations and understudied proteins, with broad implications for advancing cancer biology and informing therapeutic strategies.
Collapse
Affiliation(s)
- Zhiao Shi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - John M Elizarraras
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
36
|
Pevzner K, Simchi N, Arad G, Seger E. Identification of Protein Kinase Drug Targets Using Activity Estimation in Clinical Phosphoproteomics. Methods Mol Biol 2025; 2905:163-169. [PMID: 40163304 DOI: 10.1007/978-1-0716-4418-8_10] [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: 04/02/2025]
Abstract
This chapter describes protein kinase (will be termed here kinase) activity estimation methods and their application to clinical cancer phosphoproteomics datasets, proposing a novel approach for identification of protein kinases as therapeutic targets. Despite significant advances in genomics-based target identification, clinical proteomics and phosphoproteomics remain underutilized. We highlight the growing availability of proteomics data from projects like Clinical Proteomic Tumor Analysis Consortium (CPTAC) and Proteomics Identifications Database (PRIDE), and review key kinase activity estimation algorithms, including PTM-SEA, KSEA, Rokai, KStar, and Kinome Atlas. Applying these methods on clinical phosphoproteomic data, we demonstrate the identification of hyperactivated kinases in specific cancer indications and highlight HER2 and EGFR as benchmarks. Our description underscores the potential of integrating kinase activity estimation with clinical phosphoproteomics to uncover new therapeutic targets and develop precision oncology therapies.
Collapse
|
37
|
Tüshaus J, Eckert S, Schliemann M, Zhou Y, Pfeiffer P, Halves C, Fusco F, Weigel J, Hönikl L, Butenschön V, Todorova R, Rauert-Wunderlich H, The M, Rosenwald A, Heinemann V, Holch J, Steiger K, Delbridge C, Meyer B, Weichert W, Mogler C, Kuhn PH, Kuster B. Towards routine proteome profiling of FFPE tissue: insights from a 1,220-case pan-cancer study. EMBO J 2025; 44:304-329. [PMID: 39558110 PMCID: PMC11697351 DOI: 10.1038/s44318-024-00289-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Proteome profiling of formalin-fixed paraffin-embedded (FFPE) specimens has gained traction for the analysis of cancer tissue for the discovery of molecular biomarkers. However, reports so far focused on single cancer entities, comprised relatively few cases and did not assess the long-term performance of experimental workflows. In this study, we analyze 1220 tumors from six cancer entities processed over the course of three years. Key findings include the need for a new normalization method ensuring equal and reproducible sample loading for LC-MS/MS analysis across cohorts, showing that tumors can, on average, be profiled to a depth of >4000 proteins and discovering that current software fails to process such large ion mobility-based online fractionated datasets. We report the first comprehensive pan-cancer proteome expression resource for FFPE material comprising 11,000 proteins which is of immediate utility to the scientific community, and can be explored via a web resource. It enables a range of analyses including quantitative comparisons of proteins between patients and cohorts, the discovery of protein fingerprints representing the tissue of origin or proteins enriched in certain cancer entities.
Collapse
Affiliation(s)
- Johanna Tüshaus
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Stephan Eckert
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between DKFZ and University Center Technical University of Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Marius Schliemann
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Yuxiang Zhou
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between DKFZ and University Center Technical University of Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Pauline Pfeiffer
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Christiane Halves
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Federico Fusco
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Johannes Weigel
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Lisa Hönikl
- Department of Neurosurgery, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Vicki Butenschön
- Department of Neurosurgery, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Rumyana Todorova
- Department of Medicine III and Comprehensive Cancer Center Munich, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | | | - Matthew The
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | | | - Volker Heinemann
- Department of Medicine III and Comprehensive Cancer Center Munich, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Julian Holch
- Department of Medicine III and Comprehensive Cancer Center Munich, University Hospital, Ludwig-Maximilians University Munich, Munich, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Claire Delbridge
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Bernhard Meyer
- Department of Neurosurgery, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Wilko Weichert
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between DKFZ and University Center Technical University of Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Carolin Mogler
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between DKFZ and University Center Technical University of Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Peer-Hendrik Kuhn
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between DKFZ and University Center Technical University of Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Pathology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Bernhard Kuster
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, a Partnership between DKFZ and University Center Technical University of Munich, Munich, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
| |
Collapse
|
38
|
Rho SB, Kim BR, Lee SH, Lee CH. Translationally Controlled Tumor Protein Enhances Angiogenesis in Ovarian Tumors by Activating Vascular Endothelial Growth Factor Receptor 2 Signaling. Biomol Ther (Seoul) 2025; 33:193-202. [PMID: 39664017 PMCID: PMC11704413 DOI: 10.4062/biomolther.2024.206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
Translationally controlled tumor protein (TCTP) is a regulatory protein that plays pivotal roles in cellular processes including the cell cycle, apoptosis, microtubule stabilization, embryo development, stress responses, and cancer. However, the molecular mechanism by which it promotes tumor angiogenesis is still unclear. In this study, we explored the mechanisms underlying stimulation of angiogenesis by a novel TCTP. Recombinant TCTP enhanced vascular endothelial growth factor (VEGF)-induced endothelial cell migration, capillary-like tubular structure formation, and cell proliferation by interacting with VEGF receptor 2 (VEGFR-2) in vitro. In contrast, we showed that TCTP knockdown (using short interfering [si]TCTP) led to a decrease in ovarian tumor cells. We also examined the expression of VEGF and hypoxia inducible factor 1 (HIF-1α), an important angiogenic factor. The expression of VEGF as well as HIF-1α was dramatically decreased by siTCTP. Mechanistically, siTCTP inhibited VEGFR-2 tyrosine phosphorylation and phosphorylation of its downstream targets PI3K, Akt, and mTOR. Collectively, these findings indicate that TCTP can promote proliferation and angiogenesis via the VEGFR-2/PI3K and mTOR signaling pathways in ovarian tumor cells, providing new insight into the mechanism behind the involvement of TCTP in tumor angiogenesis.
Collapse
Affiliation(s)
- Seung Bae Rho
- Division of Cancer Biology, Research Institute, National Cancer Center, Goyang 10408, Republic of Korea
| | - Boh-Ram Kim
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea
| | - Seung-Hoon Lee
- Department of Life Science, Yong In University, Yongin 17092, Republic of Korea
| | - Chang Hoon Lee
- College of Pharmacy, Dongguk University, Goyang 10326, Republic of Korea
| |
Collapse
|
39
|
Creighton CJ. An Overview of Analytical Approaches to Cancer Proteogenomics. Methods Mol Biol 2025; 2921:93-118. [PMID: 40515986 DOI: 10.1007/978-1-0716-4502-4_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2025]
Abstract
The molecular landscape of human cancers involves multiple omics layers of complexity, from genome to proteome and beyond. Cancer proteogenomics involves the integration of protein expression patterns with somatic DNA alterations. Recently, advances in mass spectrometry-based proteomic profiling technologies have enabled the generation of combined proteomic and multi-omic data for thousands of human tumors across dozens of studies. These data in the public domain can be utilized to give us a more complete picture of cancer-specific pathways and processes and identify gene candidates for therapeutic targeting. Many proteogenomic studies are ongoing involving various cancer types according to tissue or cell of origin, including studies to predict response to therapy. In addition, pan-cancer analyses across multiple studies can identify molecular commonalities, differences, and emergent themes across tumor lineages. Data integration can determine which gene alterations at the transcriptome level are translated to the protein level. A wealth of knowledge and analytical approaches developed historically to integrate gene transcription with genomic data can be readily applied to proteogenomic analyses. Here is provided an overview of higher-level analyses of proteogenomic datasets. Such analyses include defining proteomic subtypes of cancer, exploring the impact of somatic mutations and epigenetic modifications on protein expression, cataloging proteomic correlates of more aggressive disease or drug response, and identifying enriched pathways.
Collapse
Affiliation(s)
- Chad J Creighton
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA.
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
40
|
Marino GB, Deng EZ, Clarke DJB, Diamant I, Resnick AC, Ma W, Wang P, Ma'ayan A. Protocol for using Multiomics2Targets to identify targets and driver kinases for cancer cohorts profiled with multi-omics assays. STAR Protoc 2024; 5:103457. [PMID: 39565691 PMCID: PMC11617449 DOI: 10.1016/j.xpro.2024.103457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/27/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024] Open
Abstract
The availability of multi-omics data applied to profile cancer cohorts is rapidly increasing. Here, we present a protocol for Multiomics2Targets, a computational pipeline that can identify driver cell signaling pathways, protein kinases, and cell-surface targets for immunotherapy. We describe steps for preparing the data, uploading files, and tuning parameters. We then detail procedures for running the workflow, visualizing the results, and exporting and sharing reports containing the analysis. For complete details on the use and execution of this protocol, please refer to Deng et al.1.
Collapse
Affiliation(s)
- Giacomo B Marino
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Eden Z Deng
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Daniel J B Clarke
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Ido Diamant
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA
| | - Adam C Resnick
- Department of Genetics and Genomic Sciences, Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1498, New York, NY 10029, USA
| | - Weiping Ma
- Center for Data Driven Discovery in Biomedicine, Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Pei Wang
- Center for Data Driven Discovery in Biomedicine, Division of Neurosurgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Avi Ma'ayan
- Department of Pharmacological Sciences, Mount Sinai Center for Bioinformatics, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| |
Collapse
|
41
|
Rawal O, Turhan B, Peradejordi IF, Chandrasekar S, Kalayci S, Gnjatic S, Johnson J, Bouhaddou M, Gümüş ZH. PhosNetVis: A web-based tool for fast kinase-substrate enrichment analysis and interactive 2D/3D network visualizations of phosphoproteomics data. ARXIV 2024:arXiv:2402.05016v4. [PMID: 39010877 PMCID: PMC11247916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Protein phosphorylation involves the reversible modification of a protein (substrate) residue by another protein (kinase). Liquid chromatography-mass spectrometry studies are rapidly generating massive protein phosphorylation datasets across multiple conditions. Researchers then must infer kinases responsible for changes in phosphosites of each substrate. However, tools that infer kinase-substrate interactions (KSIs) are not optimized to interactively explore the resulting large and complex networks, significant phosphosites, and states. There is thus an unmet need for a tool that facilitates user-friendly analysis, interactive exploration, visualization, and communication of phosphoproteomics datasets. We present PhosNetVis, a web-based tool for researchers of all computational skill levels to easily infer, generate and interactively explore KSI networks in 2D or 3D by streamlining phosphoproteomics data analysis steps within a single tool. PhostNetVis lowers barriers for researchers in rapidly generating high-quality visualizations to gain biological insights from their phosphoproteomics datasets. It is available at: https://gumuslab.github.io/PhosNetVis/.
Collapse
Affiliation(s)
- Osho Rawal
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- These authors contributed equally
| | - Berk Turhan
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Türkiye
- These authors contributed equally
| | - Irene Font Peradejordi
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cornell Tech, Cornell University, New York, NY 10044, USA
| | - Shreya Chandrasekar
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Cornell Tech, Cornell University, New York, NY 10044, USA
| | - Selim Kalayci
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sacha Gnjatic
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeffrey Johnson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mehdi Bouhaddou
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Zeynep H. Gümüş
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Lead contact
| |
Collapse
|
42
|
Yu Z, Song Y, Wang J, Wu Y, Wang H, Liu S, Zhu Y. Comprehensive analysis of PDE2A: a novel biomarker for prognostic value and immunotherapeutic potential in human cancers. Braz J Med Biol Res 2024; 57:e14220. [PMID: 39699377 DOI: 10.1590/1414-431x2024e14220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 10/28/2024] [Indexed: 12/20/2024] Open
Abstract
Phosphodiesterase 2A (PDE2A) plays a pivotal role in modulating cyclic nucleotide metabolism. Recent studies have shown that PDE2A is associated with some tumors, but its expression profiles, prognostic significance, and immunological roles in diverse cancer types remain unclear. Utilizing advanced bioinformatics tools, we performed a comprehensive analysis of PDE2A gene expression in multiple human cancers. Our study revealed that PDE2A expression was significantly reduced in the majority of cancer types and clinicopathological stages (I to IV) compared to normal tissues. Additionally, PDE2A expression was closely related to the prognosis of cancers such as stomach adenocarcinoma (STAD), ovarian serous cystadenocarcinoma (OV), and liver hepatocellular carcinoma (LIHC). Cox regression analyses indicated that PDE2A can act as an independent prognostic factor for these cancers. The level of PDE2A DNA methylation was significantly decreased in most cancers. Genetic alterations in PDE2A predominantly manifest in the form of amplifications. Moreover, infiltrating cells and immune checkpoint genes, including PDCD1, exhibited notable correlations with PDE2A expression. Significant associations were observed between PDE2A expression and tumor mutation burden as well as microsatellite instability. Single cell sequencing revealed PDE2A's crucial role in regulating differentiation and angiogenesis of cancer cells. Functional enrichment analysis emphasized the important role of PDE2A in synaptic transmission and tumor development. Aberrant expression of PDE2A influenced the sensitivity of various antitumor and chemotherapy drugs. This research provided a comprehensive analysis of PDE2A in human cancers, highlighting its potential as both a prognostic marker and an immunotherapy target for future research.
Collapse
Affiliation(s)
- Zhen Yu
- Nankai University Affinity the Third Central Hospital, Tianjin Third Central Hospital, Tianjin, China
| | - Yawen Song
- Nankai University Affinity the Third Central Hospital, Tianjin Third Central Hospital, Tianjin, China
| | - Jin Wang
- Nankai University Affinity the Third Central Hospital, Tianjin Third Central Hospital, Tianjin, China
- Qingpu Branch of Zhongshan Hospital Affiliated to Fudan University, Shanghai, China
| | - Yujing Wu
- Nankai University Affinity the Third Central Hospital, Tianjin Third Central Hospital, Tianjin, China
| | - Hefang Wang
- College of Chemistry, Nankai University, Tianjin, China
| | - Shuye Liu
- Nankai University Affinity the Third Central Hospital, Tianjin Third Central Hospital, Tianjin, China
| | - Yu Zhu
- Nankai University Affinity the Third Central Hospital, Tianjin Third Central Hospital, Tianjin, China
| |
Collapse
|
43
|
Teodoro L, Carreira ACO, Sogayar MC. Exploring the Complexity of Pan-Cancer: Gene Convergences and in silico Analyses. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:913-934. [PMID: 39691553 PMCID: PMC11651076 DOI: 10.2147/bctt.s489246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024]
Abstract
Cancer is a complex and multifaceted group of diseases characterized by highly intricate mechanisms of tumorigenesis and tumor progression, which complicates diagnosis, prognosis, and treatment. In recent years, targeted therapies have gained prominence by focusing on specific mutations and molecular features unique to each tumor type, offering more effective and personalized treatment options. However, it is equally critical to explore the genetic commonalities across different types of cancer, which has led to the rise of pan-cancer studies. These approaches help identify shared therapeutic targets across various tumor types, enabling the development of broader and potentially more widely applicable treatment strategies. This review aims to provide a comprehensive overview of key concepts related to tumors, including tumorigenesis processes, the tumor microenvironment, and the role of extracellular vesicles in tumor biology. Additionally, we explore the molecular interactions and mechanisms driving tumor progression, with a particular focus on the pan-cancer perspective. To achieve this, we conducted an in silico analysis using publicly available datasets, which facilitated the identification of both common and divergent genetic and molecular patterns across different tumor types. By integrating these diverse areas, this review offers a clearer and deeper understanding of the factors influencing tumorigenesis and highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Leandro Teodoro
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| | - Ana Claudia O Carreira
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Center of Human and Natural Sciences, Federal University of ABC, Santo André, São Paulo, 09280-560, Brazil
| | - Mari C Sogayar
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| |
Collapse
|
44
|
Tambi R, Zehra B, Vijayakumar A, Satsangi D, Uddin M, Berdiev BK. Artificial intelligence and omics in malignant gliomas. Physiol Genomics 2024; 56:876-895. [PMID: 39437552 DOI: 10.1152/physiolgenomics.00011.2024] [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: 02/01/2024] [Revised: 09/04/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and aggressive type of malignant glioma with an average survival time of 12-18 mo. Despite the utilization of extensive surgical resections using cutting-edge neuroimaging, and advanced chemotherapy and radiotherapy, the prognosis remains unfavorable. The heterogeneity of GBM and the presence of the blood-brain barrier further complicate the therapeutic process. It is crucial to adopt a multifaceted approach in GBM research to understand its biology and advance toward effective treatments. In particular, omics research, which primarily includes genomics, transcriptomics, proteomics, and epigenomics, helps us understand how GBM develops, finds biomarkers, and discovers new therapeutic targets. The availability of large-scale multiomics data requires the development of computational models to infer valuable biological insights for the implementation of precision medicine. Artificial intelligence (AI) refers to a host of computational algorithms that is becoming a major tool capable of integrating large omics databases. Although the application of AI tools in GBM-omics is currently in its early stages, a thorough exploration of AI utilization to uncover different aspects of GBM (subtype classification, prognosis, and survival) would have a significant impact on both researchers and clinicians. Here, we aim to review and provide database resources of different AI-based techniques that have been used to study GBM pathogenesis using multiomics data over the past decade. We summarize different types of GBM-related omics resources that can be used to develop AI models. Furthermore, we explore various AI tools that have been developed using either individual or integrated multiomics data, highlighting their applications and limitations in the context of advancing GBM research and treatment.
Collapse
Affiliation(s)
- Richa Tambi
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Binte Zehra
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Aswathy Vijayakumar
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Dharana Satsangi
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Mohammed Uddin
- Center for Applied and Translational Genomics (CATG), Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- GenomeArc Inc., Mississauga, Ontario, Canada
| | - Bakhrom K Berdiev
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- GenomeArc Inc., Mississauga, Ontario, Canada
| |
Collapse
|
45
|
Yi X, Zhao H, Hu S, Dong L, Dou Y, Li J, Gao Q, Zhang B. Tumor-associated antigen prediction using a single-sample gene expression state inference algorithm. CELL REPORTS METHODS 2024; 4:100906. [PMID: 39561714 PMCID: PMC11705763 DOI: 10.1016/j.crmeth.2024.100906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/13/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024]
Abstract
We developed a Bayesian-based algorithm to infer gene expression states in individual samples and incorporated it into a workflow to identify tumor-associated antigens (TAAs) across 33 cancer types using RNA sequencing (RNA-seq) data from the Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA). Our analysis identified 212 candidate TAAs, with 78 validated in independent RNA-seq datasets spanning seven cancer types. Eighteen of these TAAs were further corroborated by proteomics data, including 10 linked to liver cancer. We predicted that 38 peptides derived from these 10 TAAs would bind strongly to HLA-A02, the most common HLA allele. Experimental validation confirmed significant binding affinity and immunogenicity for 21 of these peptides. Notably, approximately 64% of liver tumors expressed one or more TAAs associated with these 21 peptides, positioning them as promising candidates for liver cancer therapies, such as peptide vaccines or T cell receptor (TCR)-T cell treatments. This study highlights the power of integrating computational and experimental approaches to discover TAAs for immunotherapy.
Collapse
Affiliation(s)
- Xinpei Yi
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China; Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hongwei Zhao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Shunjie Hu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Liangqing Dong
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | - Yongchao Dou
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jing Li
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital and Key Laboratory of Carcinogenesis and Cancer Invasion of the Ministry of China, Fudan University, 180 Fenglin Road, Shanghai 200032, China.
| | - Bing Zhang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
46
|
Campeanu IJ, Jiang Y, Afisllari H, Dzinic S, Polin L, Yang ZQ. Multi-omics analysis reveals CMTR1 upregulation in cancer and roles in ribosomal protein gene expression and tumor growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.621171. [PMID: 39553963 PMCID: PMC11565914 DOI: 10.1101/2024.10.30.621171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The mRNA cap methyltransferase CMTR1 plays a crucial role in RNA metabolism and gene expression regulation, yet its significance in cancer remains largely unexplored. Here, we present a comprehensive multi-omics analysis of CMTR1 across various human cancers, revealing its widespread upregulation and potential as a therapeutic target. Integrating transcriptomic and proteomic data from a large set of cancer samples, we demonstrate that CMTR1 is upregulated at the mRNA, protein, and phosphoprotein levels across multiple cancer types. Functional studies using CRISPR-mediated knockout and siRNA knockdown in breast cancer models show that CMTR1 depletion significantly inhibits tumor growth both in vitro and in vivo . Transcriptomic analysis reveals that CMTR1 primarily regulates ribosomal protein genes and other transcripts containing 5' Terminal Oligopyrimidine (TOP) motifs. Additionally, CMTR1 affects the expression of snoRNA host genes and snoRNAs, suggesting a broader role in RNA metabolism. Mechanistically, we propose that CMTR1's target specificity is partly determined by mRNA structure, particularly the presence of 5'TOP motifs. Furthermore, we identify a novel CMTR1 inhibitor, N97911, through in silico screening and biochemical assays, which demonstrates significant anti-tumor activity in vitro . Our findings establish CMTR1 as a key player in cancer biology, regulating critical aspects of RNA metabolism and ribosome biogenesis, and highlight its potential as a therapeutic target across multiple cancer types.
Collapse
|
47
|
Chakraborty D, Romero R, Rajalingam K. Integrate and conquer: pan-cancer proteogenomics uncovers cancer vulnerabilities and therapeutic opportunities. Signal Transduct Target Ther 2024; 9:289. [PMID: 39406750 PMCID: PMC11480369 DOI: 10.1038/s41392-024-02009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/16/2024] [Accepted: 10/04/2024] [Indexed: 10/19/2024] Open
Affiliation(s)
| | - Rossana Romero
- Cell Biology Unit, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
| | | |
Collapse
|
48
|
Xie Z, Lin H, Huang Y, Wang X, Lin H, Xu M, Wu J, Wu Y, Shen H, Zhang Q, Chen J, Deng Y, Xu Z, Chen Z, Lin Y, Han Y, Lin L, Yan L, Li Q, Lin X, Chi P. BAP1-mediated MAFF deubiquitylation regulates tumor growth and is associated with adverse outcomes in colorectal cancer. Eur J Cancer 2024; 210:114278. [PMID: 39151323 DOI: 10.1016/j.ejca.2024.114278] [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: 02/08/2024] [Revised: 07/14/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND Despite improvements in colorectal cancer (CRC) treatment, the prognosis for advanced CRC patients remains poor. Disruption of protein stability is one of the important factors in cancer development and progression. In this study, we aim to identify and analyze novel dysregulated proteins in CRC, assessing their significance and the mechanisms. METHODS Using quantitative proteomics, expression pattern analysis, and gain-of-function/loss-of-function experiments, we identify novel functional protein dysregulated by ubiquitin-proteasome axis in CRC. Prognostic significance was evaluated in a training cohort of 546 patients and externally validated in 794 patients. Mechanistic insights are gained through molecular biology experiments, deubiquitinating enzymes (DUBs) expression library screening, and RNA sequencing. RESULTS MAFF protein emerged as the top novel candidate substrate regulated by ubiquitin-proteasome in CRC. MAFF protein was preferentially downregulated in CRC compared to adjacent normal tissues. More importantly, multicenter cohort study identified reduced MAFF protein expression as an independent predictor of overall and disease-free survival in CRC patients. The in vitro and vivo assays showed that MAFF overexpression inhibited CRC growth, while its knockdown had the opposite effect. Intriguingly, we found the abnormal expression of MAFF protein was predominantly regulated via ubiquitination of MAFF, with K48-ubiquitin being dominant. BAP1 as a nuclear deubiquitinating enzyme (DUB), bound to and deubiquitinated MAFF, thereby stabilizing it. Such stabilization upregulated DUSP5 expression, resulting in the inhibition of ERK phosphorylation. CONCLUSIONS This study describes a novel BAP1-MAFF signaling axis which is crucial for CRC growth, potentially serving as a therapeutic target and a promising prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Zhongdong Xie
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Hanbin Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Ying Huang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaojie Wang
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Hongyue Lin
- Department of General Surgery, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Meifang Xu
- Department of Pathology, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Jiashu Wu
- Department of Science and Technology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yuecheng Wu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Hao Shen
- Department of Navy Environmental and Occupational Health, Naval Medical University, Shanghai, China
| | - Qiongying Zhang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinhua Chen
- Follow up Center, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yu Deng
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Zongbin Xu
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Zhiping Chen
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yu Lin
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Yuting Han
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Lin Lin
- Department of Pathology, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Linzhu Yan
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Qingyun Li
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China
| | - Xinjian Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China.
| | - Pan Chi
- Department of Colorectal Surgery, Union Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
49
|
Thangudu RR, Holck M, Singhal D, Pilozzi A, Edwards N, Rudnick PA, Domagalski MJ, Chilappagari P, Ma L, Xin Y, Le T, Nyce K, Chaudhary R, Ketchum KA, Maurais A, Connolly B, Riffle M, Chambers MC, MacLean B, MacCoss MJ, McGarvey PB, Basu A, Otridge J, Casas-Silva E, Venkatachari S, Rodriguez H, Zhang X. NCI's Proteomic Data Commons: A Cloud-Based Proteomics Repository Empowering Comprehensive Cancer Analysis through Cross-Referencing with Genomic and Imaging Data. CANCER RESEARCH COMMUNICATIONS 2024; 4:2480-2488. [PMID: 39225545 PMCID: PMC11413857 DOI: 10.1158/2767-9764.crc-24-0243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/22/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Proteomics has emerged as a powerful tool for studying cancer biology, developing diagnostics, and therapies. With the continuous improvement and widespread availability of high-throughput proteomic technologies, the generation of large-scale proteomic data has become more common in cancer research, and there is a growing need for resources that support the sharing and integration of multi-omics datasets. Such datasets require extensive metadata including clinical, biospecimen, and experimental and workflow annotations that are crucial for data interpretation and reanalysis. The need to integrate, analyze, and share these data has led to the development of NCI's Proteomic Data Commons (PDC), accessible at https://pdc.cancer.gov. As a specialized repository within the NCI Cancer Research Data Commons (CRDC), PDC enables researchers to locate and analyze proteomic data from various cancer types and connect with genomic and imaging data available for the same samples in other CRDC nodes. Presently, PDC houses annotated data from more than 160 datasets across 19 cancer types, generated by several large-scale cancer research programs with cohort sizes exceeding 100 samples (tumor and associated normal when available). In this article, we review the current state of PDC in cancer research, discuss the opportunities and challenges associated with data sharing in proteomics, and propose future directions for the resource. SIGNIFICANCE The Proteomic Data Commons (PDC) plays a crucial role in advancing cancer research by providing a centralized repository of high-quality cancer proteomic data, enriched with extensive clinical annotations. By integrating and cross-referencing with complementary genomic and imaging data, the PDC facilitates multi-omics analyses, driving comprehensive insights, and accelerating discoveries across various cancer types.
Collapse
Affiliation(s)
| | | | | | | | - Nathan Edwards
- Georgetown University, Washington, District of Columbia.
| | | | | | | | - Lei Ma
- ICF, Rockville, Maryland.
| | - Yi Xin
- ICF, Rockville, Maryland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Esmeralda Casas-Silva
- Center for Biomedical Informatics & Information Technology, National Cancer Institute, Rockville, Maryland.
| | | | - Henry Rodriguez
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, Maryland.
| | - Xu Zhang
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, Maryland.
| |
Collapse
|
50
|
Jiang A, Liu W, Liu Y, Hu J, Zhu B, Fang Y, Zhao X, Qu L, Lu J, Liu B, Qi L, Cai C, Luo P, Wang L. DCS, a novel classifier system based on disulfidptosis reveals tumor microenvironment heterogeneity and guides frontline therapy for clear cell renal carcinoma. JOURNAL OF THE NATIONAL CANCER CENTER 2024; 4:263-279. [PMID: 39281723 PMCID: PMC11401502 DOI: 10.1016/j.jncc.2024.06.003] [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: 05/29/2023] [Revised: 04/26/2024] [Accepted: 06/13/2024] [Indexed: 09/18/2024] Open
Abstract
Background Emerging evidence suggests that cell deaths are involved in tumorigenesis and progression, which may be treated as a novel direction of cancers. Recently, a novel type of programmed cell death, disulfidptosis, was discovered. However, the detailed biological and clinical impact of disulfidptosis and related regulators remains largely unknown. Methods In this work, we first enrolled pancancer datasets and performed multi-omics analysis, including gene expression, DNA methylation, copy number variation and single nucleic variation profiles. Then we deciphered the biological implication of disulfidptosis in clear cell renal cell carcinoma (ccRCC) by machine learning. Finally, a novel agent targeting at disulfidptosis in ccRCC was identified and verified. Results We found that disulfidptosis regulators were dysregulated among cancers, which could be explained by aberrant DNA methylation and genomic mutation events. Disulfidptosis scores were depressed among cancers and negatively correlated with epithelial mesenchymal transition. Disulfidptosis regulators could satisfactorily stratify risk subgroups in ccRCC, and a novel subtype, DCS3, owning with disulfidptosis depression, insensitivity to immune therapy and aberrant genome instability were identified and verified. Moreover, treating DCS3 with NU1025 could significantly inhibit ccRCC malignancy. Conclusion This work provided a better understanding of disulfidptosis in cancers and new insights into individual management based on disulfidptosis.
Collapse
Affiliation(s)
- Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Ying Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Junyi Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baohua Zhu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yu Fang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xuenan Zhao
- Center for Translational Medicine, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Le Qu
- Department of Urology, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Juan Lu
- Vocational Education Center, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Bing Liu
- Department of Urology, The Third Affiliated Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Lin Qi
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Changsha, China
| | - Chen Cai
- Department of Special Clinic, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|