51
|
Ainatzi S, Kaufmann SV, Silbern I, Georgiev SV, Lorenz S, Rizzoli SO, Urlaub H. Ca 2+-Triggered (de)ubiquitination Events in Synapses. Mol Cell Proteomics 2025; 24:100946. [PMID: 40089065 PMCID: PMC12008530 DOI: 10.1016/j.mcpro.2025.100946] [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: 09/06/2024] [Revised: 01/29/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025] Open
Abstract
Neuronal communication relies on neurotransmitter release from synaptic vesicles (SVs), whose dynamics are controlled by Ca2+-dependent pathways, as many thoroughly studied phosphorylation cascades. However, little is known about other post-translational modifications, such as ubiquitination. To address this, we analyzed resting and stimulated synaptosomes (isolated synapses) by quantitative mass spectrometry. We identified more than 5000 ubiquitination sites on ∼2000 proteins, the majority of which participate in SV recycling processes. Several proteins showed significant changes in ubiquitination in response to Ca2+ influx, with the most pronounced changes in CaMKIIα and the clathrin adaptor protein AP180. To validate this finding, we generated a CaMKIIα mutant lacking the ubiquitination target site (K291) and analyzed it both in neurons and non-neuronal cells. K291 ubiquitination, close to an important site for CaMKIIα autophosphorylation (T286), influences the synaptic function of this kinase. We suggest that ubiquitination in response to synaptic activity is an important regulator of synaptic function.
Collapse
Affiliation(s)
- Sofia Ainatzi
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany; Bioanalytics, Institute of Clinical Chemistry, University Medical Center, Goettingen, Germany
| | - Svenja V Kaufmann
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany; Bioanalytics, Institute of Clinical Chemistry, University Medical Center, Goettingen, Germany
| | - Ivan Silbern
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany; Bioanalytics, Institute of Clinical Chemistry, University Medical Center, Goettingen, Germany
| | - Svilen V Georgiev
- Department of Neuro- and Sensory Physiology, University Medical Center, Goettingen, Germany
| | - Sonja Lorenz
- Ubiquitin Signaling Specificity, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany
| | - Silvio O Rizzoli
- Department of Neuro- and Sensory Physiology, University Medical Center, Goettingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry, Max Planck Institute for Multidisciplinary Sciences, Goettingen, Germany; Bioanalytics, Institute of Clinical Chemistry, University Medical Center, Goettingen, Germany; Cluster of Excellence Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells (MBExC), University of Göttingen, Germany; Göttingen Center for Molecular Biosciences, Georg August University Göttingen, Germany.
| |
Collapse
|
52
|
Cormican JA, Medfai L, Wawrzyniuk M, Pašen M, Afrache H, Fourny C, Khan S, Gneiße P, Soh WT, Timelli A, Nolfi E, Pannekoek Y, Cope A, Urlaub H, Sijts AJAM, Mishto M, Liepe J. PEPSeek-Mediated Identification of Novel Epitopes From Viral and Bacterial Pathogens and the Impact on Host Cell Immunopeptidomes. Mol Cell Proteomics 2025; 24:100937. [PMID: 40044041 PMCID: PMC12002930 DOI: 10.1016/j.mcpro.2025.100937] [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: 11/27/2024] [Revised: 02/11/2025] [Accepted: 03/02/2025] [Indexed: 04/07/2025] Open
Abstract
Here, we develop PEPSeek, a web-server-based software to allow higher performance in the identification of pathogen-derived epitope candidates detected via mass spectrometry in MHC class I immunopeptidomes. We apply it to human and mouse cell lines infected with SARS-CoV-2, Listeria monocytogenes, or Chlamydia trachomatis, thereby identifying a large number of novel antigens and epitopes that we prove to be recognized by CD8+ T cells. In infected cells, we identified antigenic peptide features that suggested how the processing and presentation of pathogenic antigens differ between pathogens. The quantitative tools of PEPSeek also helped to define how C. trachomatis infection cycle could impact the antigenic landscape of the host human cell system, likely reflecting metabolic changes that occurred in the infected cells.
Collapse
Affiliation(s)
- John A Cormican
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Göttingen Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Lobna Medfai
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Magdalena Wawrzyniuk
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Martin Pašen
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Göttingen Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Hassnae Afrache
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Research group of Molecular Immunology, Francis Crick Institute, London, United Kingdom
| | - Constance Fourny
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Research group of Molecular Immunology, Francis Crick Institute, London, United Kingdom
| | - Sahil Khan
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Göttingen Graduate Center for Neurosciences, Biophysics, and Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Pascal Gneiße
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Georg-August University School of Science (GAUSS), University of Göttingen, Göttingen, Germany
| | - Wai Tuck Soh
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Arianna Timelli
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Emanuele Nolfi
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Yvonne Pannekoek
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC Location University of Amsterdam, Amsterdam Institute for Infection and Immunity, Amsterdam, The Netherlands
| | - Andrew Cope
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom; Centre for Rheumatic Diseases, King's College London, London, UK
| | - Henning Urlaub
- Research group of Bioanalytical Mass Spectrometry, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Bioanalytics, Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany; Göttingen Center for Molecular Biosciences, University of Göttingen, Göttingen, Germany
| | - Alice J A M Sijts
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands; Chair T-cell Tolerance, Leibniz Institute for Immunotherapy, Regensburg, Germany.
| | - Michele Mishto
- Centre for Inflammation Biology and Cancer Immunology, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, King's College London, London, United Kingdom; Research group of Molecular Immunology, Francis Crick Institute, London, United Kingdom.
| | - Juliane Liepe
- Research group of Quantitative and Systems Biology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany; Facility for Data Sciences and Biostatistics, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
53
|
Rippe C, Bastrup JA, Holmberg J, Kawka K, Arévalo Martinez M, Albinsson S, Jepps TA, Swärd K. Declining activity of serum response factor in aging aorta in relation to aneurysm progression. J Biol Chem 2025; 301:108400. [PMID: 40081573 PMCID: PMC12002835 DOI: 10.1016/j.jbc.2025.108400] [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: 01/31/2025] [Accepted: 03/05/2025] [Indexed: 03/16/2025] Open
Abstract
Age is a critical determinant of arterial disease, including aneurysm formation. Here, to understand the impact of aging on the arterial transcriptome, we leveraged RNA-sequencing data to define transcripts that change with advancing age in human arteries. Among the most repressed transcripts in aged individuals were those that are relevant for actomyosin structure and organization, including both myosin light chain kinase (MYLK) and smooth muscle γ-actin (ACTG2). This was associated with a reduction of serum response factor (SRF), which controls these transcripts via defined promoter elements. To determine the consequences of isolated Srf depletion, we conditionally deleted Srf in vascular smooth muscle of young mice (i8-SRF-KO mice). This led to a reduction of the SRF regulon, including Mylk and Actg2, and impaired arterial contractility, but left endothelial-dependent dilatation unaffected. Srf-depletion also increased aortic diameter and Alcian blue staining of the aortic media, which are cardinal features of aortopathy, such as aortic aneurysmal disease. Despite this, i8-SRF-KO mice were protected from aortic lesions elicited by angiotensin II (AngII). Proteomics demonstrated that Srf-depletion mimicked a protein signature of AngII treatment involving increases of the mechanoresponsive transcriptional coactivators YAP and TAZ and reduction of the Hippo kinase Lats2. Protection from aortopathy could be overcome by changing the order of KO induction and AngII administration resulting in advanced aneurysms in both i8-SRF-KO and control mice. Our work provides important insights into the molecular underpinnings of age-dependent changes in aortic function and mechanisms of adaptation in hypertension.
Collapse
Affiliation(s)
- Catarina Rippe
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Joakim Armstrong Bastrup
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johan Holmberg
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Katarzyna Kawka
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Sebastian Albinsson
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Karl Swärd
- Vascular Physiology Environment, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
54
|
Ma Y, Qin LY, Ding X, Wu AP. Diversity, Complexity, and Challenges of Viral Infectious Disease Data in the Big Data Era: A Comprehensive Review. CHINESE MEDICAL SCIENCES JOURNAL = CHUNG-KUO I HSUEH K'O HSUEH TSA CHIH 2025; 40:29-44. [PMID: 40165755 DOI: 10.24920/004461] [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] [Indexed: 04/02/2025]
Abstract
Viral infectious diseases, characterized by their intricate nature and wide-ranging diversity, pose substantial challenges in the domain of data management. The vast volume of data generated by these diseases, spanning from the molecular mechanisms within cells to large-scale epidemiological patterns, has surpassed the capabilities of traditional analytical methods. In the era of artificial intelligence (AI) and big data, there is an urgent necessity for the optimization of these analytical methods to more effectively handle and utilize the information. Despite the rapid accumulation of data associated with viral infections, the lack of a comprehensive framework for integrating, selecting, and analyzing these datasets has left numerous researchers uncertain about which data to select, how to access it, and how to utilize it most effectively in their research.This review endeavors to fill these gaps by exploring the multifaceted nature of viral infectious diseases and summarizing relevant data across multiple levels, from the molecular details of pathogens to broad epidemiological trends. The scope extends from the micro-scale to the macro-scale, encompassing pathogens, hosts, and vectors. In addition to data summarization, this review thoroughly investigates various dataset sources. It also traces the historical evolution of data collection in the field of viral infectious diseases, highlighting the progress achieved over time. Simultaneously, it evaluates the current limitations that impede data utilization.Furthermore, we propose strategies to surmount these challenges, focusing on the development and application of advanced computational techniques, AI-driven models, and enhanced data integration practices. By providing a comprehensive synthesis of existing knowledge, this review is designed to guide future research and contribute to more informed approaches in the surveillance, prevention, and control of viral infectious diseases, particularly within the context of the expanding big-data landscape.
Collapse
Affiliation(s)
- Yun Ma
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing 107302, China
| | - Lu-Yao Qin
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing 107302, China
| | - Xiao Ding
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing 107302, China.
| | - Ai-Ping Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Peking Union Medical College), Ministry of Education, Beijing 107302, China.
| |
Collapse
|
55
|
Maklad A, Sedeeq M, Baghaei K, Wilson R, Heath JA, Gueven N, Azimi I. Role of LIN28B in the Regulation of Ribosomal Biogenesis and Lipid Metabolism in Medulloblastoma Brain Cancer Cells. Proteomes 2025; 13:14. [PMID: 40265419 PMCID: PMC12015845 DOI: 10.3390/proteomes13020014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/07/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025] Open
Abstract
Background: Medulloblastoma (MB) is the most aggressive paediatric brain cancer, highlighting the urgent need for new diagnostic and prognostic biomarkers and improved treatments to enhance patient outcomes. Our previous study identified LIN28B, an RNA-binding protein, as a potential diagnostic and prognostic marker for MB and a pharmacological target to inhibit MB cell proliferation and stemness. However, the specific role of LIN28B and its mechanism of action in MB had not been studied. Methods: This study assessed LIN28B's role in Daoy MB cells using siRNA-mediated silencing. LIN28B silencing was achieved with Dharmacon ON-TARGETplus SMARTpool and confirmed by Western blotting. Proliferation and protein assays evaluated the cell metabolic activity and viability. A proteomics analysis was conducted to examine the effect of LIN28B knockdown on the MB cell protein expression profile. The intracellular lipid droplets were assessed using the Nile Red Staining Kit, and nucleolar B23 protein levels were assessed by immunofluorescence. Both were visualised with a high-content IN Cell Analyser 2200. Results: Effective LIN28B silencing (>80%) was achieved in each experiment. LIN28B knockdown reduced the MB cell viability, impaired ribosome biogenesis, and promoted cellular lipid accumulation, as supported by proteomics and cell-based assays. Conclusions: This study highlights LIN28B as a promising target for regulating MB cell growth, ribosomal biogenesis, and lipid metabolism.
Collapse
Affiliation(s)
- Ahmed Maklad
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS 7001, Australia; (A.M.); (M.S.); (N.G.)
| | - Mohammed Sedeeq
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS 7001, Australia; (A.M.); (M.S.); (N.G.)
| | - Kaveh Baghaei
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Melbourne, VIC 3168, Australia;
| | - Richard Wilson
- Central Science Laboratory, College of Science and Engineering, University of Tasmania, Hobart, TAS 7001, Australia;
| | - John A. Heath
- Children’s Cancer Centre, Monash Children’s Hospital, Melbourne, VIC 3168, Australia;
| | - Nuri Gueven
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS 7001, Australia; (A.M.); (M.S.); (N.G.)
| | - Iman Azimi
- School of Pharmacy and Pharmacology, College of Health and Medicine, University of Tasmania, Hobart, TAS 7001, Australia; (A.M.); (M.S.); (N.G.)
- Monash Biomedicine Discovery Institute, Department of Pharmacology, Monash University, Melbourne, VIC 3168, Australia;
| |
Collapse
|
56
|
Gorodetska I, Lukiyanchuk V, Gawin M, Sliusar M, Linge A, Lohaus F, Hölscher T, Erdmann K, Fuessel S, Borkowetz A, Wojakowska A, Fochtman D, Reardon M, Choudhury A, Antonelli Y, Leal-Egaña A, Köseer AS, Kahya U, Püschel J, Petzold A, Klusa D, Peitzsch C, Kronstein-Wiedemann R, Tonn T, Marczak L, Thomas C, Widłak P, Pietrowska M, Krause M, Dubrovska A. Blood-based detection of MMP11 as a marker of prostate cancer progression regulated by the ALDH1A1-TGF-β1 signaling mechanism. J Exp Clin Cancer Res 2025; 44:105. [PMID: 40122809 PMCID: PMC11931756 DOI: 10.1186/s13046-025-03299-6] [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: 07/25/2024] [Accepted: 01/12/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the second most common type of tumor diagnosed in men and the fifth leading cause of cancer-related death in male patients. The response of metastatic disease to standard treatment is heterogeneous. As for now, there is no curative treatment option available for metastatic PCa, and the clinical tests capable of predicting metastatic dissemination and metastatic response to the therapies are lacking. Our recent study identified aldehyde dehydrogenases ALDH1A1 and ALDH1A3 as critical regulators of PCa metastases. Still, the exact mechanisms mediating the role of these proteins in PCa metastatic dissemination remain not fully understood, and plasma-based biomarkers of these metastatic mechanisms are not available. METHODS Genetic silencing, gene overexpression, or treatment with different concentrations of the retinoic acid (RA) isomers, which are the products of ALDH catalytic activity, were used to modulate the interplay between retinoic acid receptors (RARs) and androgen receptor (AR). RNA sequencing (RNAseq), reporter gene assays, and chromatin immunoprecipitation (ChIP) analysis were employed to validate the role of RARs and AR in the regulation of the transforming growth factor-beta 1 (TGFB1) expression. Gene expression levels of ALDH1A1, ALDH1A3, and the matrix metalloproteinase 11 (MMP11) and their correlation with pathological parameters and clinical outcomes were analysed by mining several publicly available patient datasets as well as our multi-center transcriptomic dataset from patients with high-risk and locally advanced PCa. The level of MMP11 protein was analysed by enzyme-linked immunosorbent assay (ELISA) in independent cohorts of plasma samples from patients with primary or metastatic PCa and healthy donors, while plasma proteome profiles were obtained for selected subsets of PCa patients. RESULTS We could show that ALDH1A1 and ALDH1A3 genes differently regulate TGFB1 expression in a RAR- and AR-dependent manner. We further observed that the TGF-β1 pathway contributes to the regulation of the MMPs, including MMP11. We have confirmed the relevance of MMP11 as a promising clinical marker for PCa using several independent gene expression datasets. Further, we have validated plasma MMP11 level as a prognostic biomarker in patients with metastatic PCa. Finally, we proposed a hypothetical ALDH1A1/MMP11-related plasma proteome-based prognostic signature. CONCLUSIONS TGFB1/MMP11 signaling contributes to the ALDH1A1-driven PCa metastases. MMP11 is a promising blood-based biomarker of PCa progression.
Collapse
Affiliation(s)
- Ielizaveta Gorodetska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Vasyl Lukiyanchuk
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Marta Gawin
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Myroslava Sliusar
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Annett Linge
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Fabian Lohaus
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Tobias Hölscher
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Kati Erdmann
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Susanne Fuessel
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Angelika Borkowetz
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Anna Wojakowska
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
| | - Daniel Fochtman
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
| | - Mark Reardon
- Division of Cancer Sciences, Translational Radiobiology Group, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, Translational Radiobiology Group, University of Manchester, Manchester Cancer Research Centre, Christie NHS Foundation Trust, Manchester, UK
| | - Yasmin Antonelli
- Institute for Molecular Systems Engineering and Advanced Materials, Heidelberg University, Heidelberg, Germany
| | - Aldo Leal-Egaña
- Institute for Molecular Systems Engineering and Advanced Materials, Heidelberg University, Heidelberg, Germany
| | - Ayse Sedef Köseer
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Uğur Kahya
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Jakob Püschel
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
| | - Andrea Petzold
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
| | - Daria Klusa
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Claudia Peitzsch
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Romy Kronstein-Wiedemann
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Torsten Tonn
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- Experimental Transfusion Medicine, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Institute for Transfusion Medicine Dresden, German Red Cross Blood Donation Service North-East, Dresden, Germany
| | - Lukasz Marczak
- Institute of Bioorganic Chemistry Polish Academy of Sciences, Poznań, Poland
| | - Christian Thomas
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
- Department of Urology, Faculty of Medicine, University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Piotr Widłak
- 2nd Department of Radiology, Medical University of Gdansk, Gdansk, Poland
| | - Monika Pietrowska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Mechthild Krause
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany
| | - Anna Dubrovska
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Helmholtz-Zentrum Dresden -Rossendorf, Dresden, Germany.
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany, and Helmholtz Association/Helmholtz-Zentrum Dresden - Rossendorf (HZDR), Dresden, Germany.
| |
Collapse
|
57
|
Garde C, Pavlidis MA, Garces P, Lange EJ, Ramarathinam SH, Sokač M, Pandey K, Faridi P, Ahrenfeldt J, Chung S, Friis S, Kleine-Kohlbrecher D, Birkbak NJ, Kringelum JV, Rønø B, Purcell AW, Trolle T. Endogenous viral elements constitute a complementary source of antigens for personalized cancer vaccines. NPJ Vaccines 2025; 10:54. [PMID: 40113807 PMCID: PMC11926357 DOI: 10.1038/s41541-025-01107-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Personalized cancer vaccines (PCVs) largely leverage neoantigens arising from somatic mutations, limiting their application to patients with relatively high tumor mutational burden (TMB). This underscores the need for alternative antigens to design PCVs for low TMB cancers. To this end, we substantiate endogenous retroviral elements (EVEs) as tumor antigens through large-scale genomic analyses of healthy tissues and solid cancers. These analyses revealed that the breadth of EVE expression in tumors stratify checkpoint inhibitor-treated melanoma patients into groups with differential overall and progression-free survival. To enable the design of PCVs containing EVE-derived epitopes with therapeutic potential, we developed a computational pipeline, ObsERV. We show that EVE-derived peptides are presented as epitopes on tumors and can be predicted by ObsERV. Preclinical testing of ObsERV demonstrates induction of sustained poly-functional CD4+ and CD8+ T-cell responses as well as long-term tumor protection. As such, EVEs may facilitate and improve PCVs, especially for low-TMB patients.
Collapse
Affiliation(s)
- Christian Garde
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark.
| | | | - Pablo Garces
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| | - Emma J Lange
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| | - Sri H Ramarathinam
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Mateo Sokač
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Kirti Pandey
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Pouya Faridi
- Department of Medicine, School of Clinical Sciences, Monash University, Clayton, VIC, Australia
| | - Johanne Ahrenfeldt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Shanzou Chung
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Stine Friis
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| | | | - Nicolai J Birkbak
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | - Birgitte Rønø
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| | - Anthony W Purcell
- Department of Biochemistry and Molecular Biology & Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Thomas Trolle
- Evaxion Biotech A/S, Dr Neergaards Vej 5F, Hørsholm, Denmark
| |
Collapse
|
58
|
Paul S, Das S, Banerjea M, Chaudhuri S, Das B. The ATP-dependent DEAD-box RNA helicase Dbp2 regulates the glucose/nitrogen stress response in baker's yeast by modulating reversible nuclear retention and decay of SKS1 mRNA. Genetics 2025; 229:iyae221. [PMID: 39739574 DOI: 10.1093/genetics/iyae221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/23/2024] [Indexed: 01/02/2025] Open
Abstract
In Saccharomyces cerevisiae, SKS1 mRNA encoding a glucose-sensing serine/threonine kinase belongs to "nucleus-retained" (NR) mRNAs representing a subset of otherwise normal transcripts, which exhibits slow nuclear export and excessively long nuclear dwell time. Nuclear retention of the SKS1 mRNA triggered by a 202 nt "export-retarding" nuclear zip code element promotes its rapid degradation in the nucleus by the nuclear exosome/CTEXT. In this investigation, we demonstrate that Dbp2p, an ATP-dependent DEAD-box RNA helicase binds to SKS1 and other NR-mRNAs and thereby inhibits their export by antagonizing with the binding of the export factors Mex67p/Yra1p. Consistent with this observation, a significant portion of these NR-mRNAs was found to localize into the cytoplasm in a yeast strain carrying a deletion in the DBP2 gene with the concomitant enhancement of its steady-state level and stability. This observation supports the view that Dbp2p promotes the nuclear retention of NR-mRNAs to trigger their subsequent nuclear degradation. Further analysis revealed that Dbp2p-dependent nuclear retention of SKS1 mRNA is reversible, which plays a crucial role in the adaptability and viability of the yeast cells in low concentrations of glucose/nitrogen in the growth medium. At high nutrient levels when the function of Sks1p is not necessary, SKS1 mRNA is retained in the nucleus and degraded. In contrast, during low glucose/nitrogen levels when Sks1p is vital to respond to such situations, the nuclear retention of SKS1 mRNA is relieved to permit its increased nuclear export and translation leading to a huge burst of cytoplasmic Sks1p.
Collapse
Affiliation(s)
- Soumita Paul
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, India
| | - Subhadeep Das
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, India
| | - Mayukh Banerjea
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, India
| | - Shouvik Chaudhuri
- Department of Mechanical Engineering, Jadavpur University, Kolkata 700032, India
| | - Biswadip Das
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata 700032, India
| |
Collapse
|
59
|
Nishimura Y, Bittel A, Jagan A, Chen YW, Burniston J. Proteomic profiling uncovers sexual dimorphism in the muscle response to wheel running exercise in the FLExDUX4 murine model of facioscapulohumeral muscular dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.15.639012. [PMID: 40166172 PMCID: PMC11956996 DOI: 10.1101/2025.03.15.639012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
FLExDUX4 is a murine experimental model of facioscapulohumeral muscular dystrophy (FSHD) characterized by chronic, low levels of leaky expression of the human full-length double homeobox 4 gene (DUX4-fl). FLExDUX4 mice exhibit mild pathologies and functional deficits similar to people affected by FSHD. Proteomic studies in FSHD could offer new insights into disease mechanisms underpinned by post-transcriptional processes. We used mass spectrometry-based proteomics to quantify the abundance of 1322 proteins in triceps brachii muscle, encompassing both male and female mice in control and free voluntary wheel running (VWR) in Wild-type (n=3) and FLExDUX4 (n=3) genotypes. We report the triceps brachii proteome of FLExDUX4 mice recapitulates key skeletal muscle clinical characteristics of human FSHD, including alterations to mitochondria, RNA metabolism, oxidative stress, and apoptosis. RNA-binding proteins exhibit a sex-specific difference in FLExDUX4 mice. Sexual dimorphism of mitochondrial protein adaptation to exercise was uncovered specifically in FLExDUX4 mice, where females increased, but males decreased mitochondrial proteins after a 6-week of VWR. Our results highlight the importance of identifying sex-specific diagnostic biomarkers to enable more reliable monitoring of FSHD therapeutic targets. Our data provides a resource for the FSHD research community to explore the burgeoning aspect of sexual dimorphism in FSHD.
Collapse
Affiliation(s)
- Yusuke Nishimura
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, L3 3AF, United Kingdom
| | - Adam Bittel
- Center for Genetic Medicine Research, Children’s National Medical Center, Washington, DC, USA
| | - Abhishek Jagan
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, L3 3AF, United Kingdom
| | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children’s National Medical Center, Washington, DC, USA
| | - Jatin Burniston
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, L3 3AF, United Kingdom
| |
Collapse
|
60
|
Rega C, Tsitsa I, Roumeliotis TI, Krystkowiak I, Portillo M, Yu L, Vorhauser J, Pines J, Mansfeld J, Choudhary J, Davey NE. High resolution profiling of cell cycle-dependent protein and phosphorylation abundance changes in non-transformed cells. Nat Commun 2025; 16:2579. [PMID: 40089461 PMCID: PMC11910661 DOI: 10.1038/s41467-025-57537-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 02/24/2025] [Indexed: 03/17/2025] Open
Abstract
The cell cycle governs a precise series of molecular events, regulated by coordinated changes in protein and phosphorylation abundance, that culminates in the generation of two daughter cells. Here, we present a proteomic and phosphoproteomic analysis of the human cell cycle in hTERT-RPE-1 cells using deep quantitative mass spectrometry by isobaric labelling. By analysing non-transformed cells and improving the temporal resolution and coverage of key cell cycle regulators, we present a dataset of cell cycle-dependent protein and phosphorylation site oscillation that offers a foundational reference for investigating cell cycle regulation. These data reveal regulatory intricacies including proteins and phosphorylation sites exhibiting cell cycle-dependent oscillation, and proteins targeted for degradation during mitotic exit. Integrated with complementary resources, our data link cycle-dependent abundance dynamics to functional changes and are accessible through the Cell Cycle database (CCdb), an interactive web-based resource for the cell cycle community.
Collapse
Affiliation(s)
- Camilla Rega
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Ifigenia Tsitsa
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | | | | | - Maria Portillo
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Lu Yu
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Julia Vorhauser
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Jonathon Pines
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Jörg Mansfeld
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Jyoti Choudhary
- Division of Cancer Biology, The Institute of Cancer Research, London, UK
| | - Norman E Davey
- Division of Cancer Biology, The Institute of Cancer Research, London, UK.
| |
Collapse
|
61
|
Yuan Y, Iannetta AA, Kim M, Sadecki PW, Arend M, Tsichla A, Águila Ruiz-Sola M, Kepesidis G, Falconet D, Thevenon E, Tardif M, Brugière S, Couté Y, Kleman JP, Sizova I, Schilling M, Jouhet J, Hegemann P, Li-Beisson Y, Nikoloski Z, Bastien O, Hicks LM, Petroutsos D. Phototropin connects blue light perception to starch metabolism in green algae. Nat Commun 2025; 16:2545. [PMID: 40087266 PMCID: PMC11909140 DOI: 10.1038/s41467-025-57809-3] [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: 04/05/2024] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
In photosynthetic organisms, light acts as an environmental signal to control their development and physiology, as well as energy source to drive the conversion of CO2 into carbohydrates used for growth or storage. The main storage carbohydrate in green algae is starch, which accumulates during the day and is broken down at night to meet cellular energy demands. The signaling role of light quality in the regulation of starch accumulation remains unexplored. Here, we identify PHOTOTROPIN-MEDIATED SIGNALING KINASE 1 (PMSK1) as a key regulator of starch metabolism in Chlamydomonas reinhardtii. In its phosphorylated form (PMSK1-P), it activates GLYCERALDEHYDE-3-PHOSPHATE DEHYDROGENASE (GAP1), promoting starch biosynthesis. We show that blue light, perceived by PHOTOTROPIN, induces PMSK1 dephosphorylation that in turn represses GAP1 mRNA levels and reduces starch accumulation. These findings reveal a previously uncharacterized blue light-mediated signaling pathway that advances our understanding of photoreceptor-controlled carbon metabolism in microalgae.
Collapse
Affiliation(s)
- Yizhong Yuan
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA, USA
| | - Anthony A Iannetta
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Minjae Kim
- Institute de Biosciences et Biotechnologies Aix-Marseille, Aix Marseille University, CEA, CNRS, BIAM, Saint Paul-Lez-Durance, France
| | - Patric W Sadecki
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Marius Arend
- Bioinformatics Department, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
- Systems Biology and Mathematical Modeling Group, Max Planck Institute of Molecular Plant Physiology, Potsdam, Germany
- Bioinformatics and Mathematical Modeling Department, Center of Plant Systems Biology and Biotechnology, Plovdiv, Bulgaria
| | - Angeliki Tsichla
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - M Águila Ruiz-Sola
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas-Universidad de Sevilla, Sevilla, Spain
| | - Georgios Kepesidis
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France
- Sandia National Laboratories, Livermore, CA, USA
| | - Denis Falconet
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France
| | - Emmanuel Thevenon
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France
| | - Marianne Tardif
- Université Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, Grenoble, France
| | - Sabine Brugière
- Université Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, Grenoble, France
| | - Yohann Couté
- Université Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, Grenoble, France
| | | | - Irina Sizova
- Institute of Biology, Experimental Biophysics, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marion Schilling
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France
| | - Juliette Jouhet
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France
| | - Peter Hegemann
- Institute of Biology, Experimental Biophysics, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yonghua Li-Beisson
- Institute de Biosciences et Biotechnologies Aix-Marseille, Aix Marseille University, CEA, CNRS, BIAM, Saint Paul-Lez-Durance, France
| | - Zoran Nikoloski
- Bioinformatics Department, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
- Systems Biology and Mathematical Modeling Group, Max Planck Institute of Molecular Plant Physiology, Potsdam, Germany
- Bioinformatics and Mathematical Modeling Department, Center of Plant Systems Biology and Biotechnology, Plovdiv, Bulgaria
| | - Olivier Bastien
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France
| | - Leslie M Hicks
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dimitris Petroutsos
- Université Grenoble Alpes, CNRS, CEA, INRAE, IRIG-LPCV, Grenoble, France.
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
62
|
Hossain MK, Unger L, Larsen U, Altankhuyag A, Legøy TA, Paulo JA, Vethe H, Ghila L. Mapping the initial effects of carcinogen-induced oncogenic transformation in the mouse bladder. Exp Cell Res 2025; 446:114452. [PMID: 39988124 DOI: 10.1016/j.yexcr.2025.114452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/31/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
Characterizing the initial stages of oncogenic transformation allows the identification of tumor-promoting processes before the inherent clonal selection of the aggressive clones. Here, we used global proteomics, genetic cell tracing, and immunofluorescence to dynamically map the very early stages of cancer initiation in a mouse model of bladder cancer. We observed a very rapid and incremental proteome dysregulation, with changes in the energy metabolism, proliferation and immune signatures dominating the landscape. The changes in the lipid metabolism were immediate and defined by an increase fatty acid metabolism and lipid transport, followed by the activation of the immune landscape. Alongside the changes in the immune signature and lipid metabolism, we also mapped a clear increase in the cell cycle-related pathways and proliferation. Proliferation was mainly restricted to the basal epithelial layer rapidly leading to urothelium thickening, despite the progressive loss of the superficial layer. Moreover, we observed a tilt in the energy balance towards increased glucose metabolism, probably characterizing cells of the tumor microenvironment. All of the observed proteome signature changes were persistent, being retained and sometimes intensified or diversified along the timeline. The signatures observed in this pilot suggest these processes as potentially targetable drivers of the future neoplastic transformations in the bladder.
Collapse
Affiliation(s)
- Md Kaykobad Hossain
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Lucas Unger
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Ulrik Larsen
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | | | - Thomas Aga Legøy
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Heidrun Vethe
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, Faculty of Medicine, University of Bergen, Norway
| | - Luiza Ghila
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Norway.
| |
Collapse
|
63
|
Vidović D, Shamsaei B, Schürer SC, Kogan P, Chojnacki S, Kouril M, Medvedovic M, Niu W, Azeloglu EU, Birtwistle MR, Chen Y, Chen T, Hansen J, Hu B, Iyengar R, Jayaraman G, Li H, Liu T, Sobie EA, Xiong Y, Berberich MJ, Bradshaw G, Chung M, Everley RA, Gaudio B, Hafner M, Kalocsay M, Mills CE, Nariya MK, Sorger PK, Subramanian K, Victor C, Banuelos M, Dardov V, Holewinski R, Manalo DM, Mandefro B, Matlock AD, Ornelas L, Sareen D, Svendsen CN, Vaibhav V, Van Eyk JE, Venkatraman V, Finkbiener S, Fraenkel E, Rothstein J, Thompson L, Asiedu J, Carr SA, Christianson KE, Davison D, Dele-Oni DO, DeRuff KC, Egri SB, Jacome ASV, Jaffe JD, Lam D, Litichevskiy L, Lu X, Mullahoo J, Officer A, Papanastasiou M, Peckner R, Toder C, Blanchard J, Bula M, Ko T, Tsai LH, Young JZ, Sharma V, Pillai A, Meller J, MacCoss MJ. Comprehensive proteomics metadata and integrative web portals facilitate sharing and integration of LINCS multiomics data. Mol Cell Proteomics 2025:100947. [PMID: 40089066 DOI: 10.1016/j.mcpro.2025.100947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 02/14/2025] [Accepted: 03/11/2025] [Indexed: 03/17/2025] Open
Abstract
The Library of Integrated Network-based Cellular Signatures (LINCS), an NIH Common Fund program, has cataloged and analyzed cellular function and molecular activity profiles in response to >80,000 perturbing agents that are potentially disruptive to cells. Because of the importance of proteins and their modifications to the response of specific cellular perturbations, four of the six LINCS centers have included significant proteomics efforts in the characterization of the resulting phenotype. This manuscript aims to describe this effort and the data harmonization and integration of the LINCS proteomics data discussed in recent LINCS papers.
Collapse
Affiliation(s)
- Dušica Vidović
- BD2K-LINCS DCIC, Department of Molecular and Cellular Pharmacology, University of Miami, Miami, FL 33146; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL 33146
| | - Behrouz Shamsaei
- BD2K-LINCS DCIC, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45220
| | - Stephan C Schürer
- BD2K-LINCS DCIC, Department of Molecular and Cellular Pharmacology, University of Miami, Miami, FL 33146; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, FL 33146; BD2K-LINCS DCIC, Institute for Data Science and Computing, University of Miami, FL 33146
| | - Phillip Kogan
- BD2K-LINCS DCIC, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45220
| | - Szymon Chojnacki
- BD2K-LINCS DCIC, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45220
| | - Michal Kouril
- BD2K-LINCS DCIC, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45220
| | - Mario Medvedovic
- BD2K-LINCS DCIC, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45220
| | - Wen Niu
- BD2K-LINCS DCIC, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45220
| | - Evren U Azeloglu
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Marc R Birtwistle
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Yibang Chen
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Tong Chen
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; DToxS, Center for Advanced Proteomics Research, Rutgers University New Jersey Medical School, Newark, NJ 07103
| | - Jens Hansen
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Bin Hu
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ravi Iyengar
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Gomathi Jayaraman
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Hong Li
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; DToxS, Center for Advanced Proteomics Research, Rutgers University New Jersey Medical School, Newark, NJ 07103
| | - Tong Liu
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029; DToxS, Center for Advanced Proteomics Research, Rutgers University New Jersey Medical School, Newark, NJ 07103
| | - Eric A Sobie
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Yuguang Xiong
- DToxS, Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | | | - Gary Bradshaw
- HMS LINCS Center, Harvard Medical School, Boston, MA 02115
| | - Mirra Chung
- HMS LINCS Center, Harvard Medical School, Boston, MA 02115
| | | | - Ben Gaudio
- HMS LINCS Center, Harvard Medical School, Boston, MA 02115
| | - Marc Hafner
- HMS LINCS Center, Harvard Medical School, Boston, MA 02115
| | | | | | | | - Peter K Sorger
- HMS LINCS Center, Harvard Medical School, Boston, MA 02115
| | | | - Chiara Victor
- HMS LINCS Center, Harvard Medical School, Boston, MA 02115
| | - Maria Banuelos
- NeuroLINCS, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Victoria Dardov
- NeuroLINCS, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | | | | | - Berhan Mandefro
- NeuroLINCS, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | | | - Loren Ornelas
- NeuroLINCS, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Dhruv Sareen
- NeuroLINCS, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | | | - Vineet Vaibhav
- NeuroLINCS, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | | | | | - Steve Finkbiener
- NeuroLINCS, Gladstone Institute of Neurological Disease and the Departments of Neurology and Physiology, University of California San Francisco, San Francisco, CA 94158
| | - Ernest Fraenkel
- NeuroLINCS, Department of Biological Engineering, MIT, Cambridge, MA 02142
| | - Jeffrey Rothstein
- NeuroLINCS, Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205
| | - Leslie Thompson
- NeuroLINCS, Departments of Psychiatry and Human Behavior and Neurobiology and Behavior, University of California Irvine, Irvine, CA 92697
| | - Jacob Asiedu
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - Steven A Carr
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | | | - Desiree Davison
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | | | | | - Shawn B Egri
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | | | - Jacob D Jaffe
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - Daniel Lam
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - Lev Litichevskiy
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - Xiaodong Lu
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - James Mullahoo
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - Adam Officer
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | | | - Ryan Peckner
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - Caidin Toder
- PCCSE, The Broad Institute of Harvard and MIT, Cambridge, MA 02142
| | - Joel Blanchard
- PCCSE, Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Michael Bula
- PCCSE, Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Tak Ko
- PCCSE, Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Li-Huei Tsai
- PCCSE, Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jennie Z Young
- PCCSE, Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Vagisha Sharma
- PCCSE, Department of Genome Sciences, University of Washington, Seattle, WA 98195
| | | | - Jarek Meller
- BD2K-LINCS DCIC, Department of Environmental and Public Health Sciences, University of Cincinnati, Cincinnati, OH 45220.
| | - Michael J MacCoss
- PCCSE, Department of Genome Sciences, University of Washington, Seattle, WA 98195.
| |
Collapse
|
64
|
Grothey B, Lyu SI, Quaas A, Simon AG, Jung JO, Schröder W, Bruns CJ, Schiffmann LM, Popp FC, Schmidt T, Knipper K. Proteomic characterization of MET-amplified esophageal adenocarcinomas reveals enrichment of alternative splicing- and androgen signaling-related proteins. Cell Mol Life Sci 2025; 82:112. [PMID: 40074836 PMCID: PMC11904063 DOI: 10.1007/s00018-025-05635-7] [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: 09/10/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Esophageal adenocarcinomas (EACs) represent an evolving tumor entity with high mortality rates. MET amplification is a recurrent driver in EACs and is associated with decreased patient survival. However, the response to MET inhibitors is limited. Recent studies have identified several mechanisms that lead to resistance against MET inhibitors in different tumor entities. Nonetheless, a characterization of additional vulnerable targets beyond MET has not been conducted in MET-amplified EACs. METHODS In this study, we determined the MET amplification status in a cohort of more than 900 EACs using fluorescence in situ hybridization (FISH) and compared the proteomes of MET-amplified (n = 20) versus non-amplified tumors (n = 39) by mass spectrometry. RESULTS We identified a phenotype, present in almost all MET-amplified tumors, which shows an enrichment of alternative RNA splicing, and androgen receptor signaling proteins, as well as decreased patient survival. Additionally, our analyses revealed a negative correlation between MET expression and patient survival in MET-amplified EACs, indicating biological heterogeneity with clinical relevance despite the presence of MET amplification as the predominant oncogenic driver. Furthermore, quantitative immunohistochemical analysis of the inflammatory tumor microenvironment showed that an increased percentage of M2 macrophages is associated with lower overall survival in MET-amplified EACs. CONCLUSIONS Our results provide valuable insights into possible new therapeutic approaches for MET-amplified EACs for further research.
Collapse
Affiliation(s)
- Bastian Grothey
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Su Ir Lyu
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Alexander Quaas
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Adrian Georg Simon
- Faculty of Medicine, Institute of Pathology, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Jin-On Jung
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Wolfgang Schröder
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Christiane J Bruns
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Lars M Schiffmann
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Felix C Popp
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Thomas Schmidt
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Karl Knipper
- Faculty of Medicine, Department of General, Visceral and Cancer Surgery, University Hospital of Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
65
|
Powell CE, Dohnalová L, Eisert RJ, Sun ZYJ, Seo HS, Dhe-Paganon S, Thaiss CA, Devlin AS. Gut Microbiome-Produced Bile Acid Metabolite Lengthens Circadian Period in Host Intestinal Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.10.642513. [PMID: 40161646 PMCID: PMC11952472 DOI: 10.1101/2025.03.10.642513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Host circadian signaling, feeding, and the gut microbiome are tightly interconnected. Changes in the gut microbial community can affect the expression of core clock genes, but the specific metabolites and molecular mechanisms that mediate this relationship remain largely unknown. Here, we sought to identify gut microbial metabolites that impact circadian signaling. Through a phenotypic screen of a focused library of gut microbial metabolites, we identified a bile acid metabolite, lithocholic acid (LCA), as a circadian modulator. LCA lengthened the circadian period of core clock gene hPer2 transcription in a dose-responsive manner in human colonic cells. We found evidence that LCA modulates the casein kinase 1 δ/ε (CK1δ/ε)-protein phosphatase 1 (PP1) feedback loop and stabilizes core clock protein cryptochrome 2 (CRY2). Furthermore, we showed that LCA feeding alters circadian transcription in mouse distal ileum and colon. Taken together, our work identifies LCA as a molecular link between host circadian biology and the microbiome. Because bile acids are secreted in response to feeding, our work provides potential mechanistic insight into the molecular nature of the food-entrainable oscillator by which peripheral clocks adapt to the timing of food intake. Given the association between circadian rhythm, feeding, and metabolic disease, our insights may offer a new avenue for modulating host health.
Collapse
|
66
|
Nicholas B, Bailey A, McCann KJ, Walker RC, Johnson P, Elliott T, Underwood TJ, Skipp P. Comparative analysis of protein expression between oesophageal adenocarcinoma and normal adjacent tissue. PLoS One 2025; 20:e0318572. [PMID: 40073002 PMCID: PMC11902292 DOI: 10.1371/journal.pone.0318572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/19/2025] [Indexed: 03/14/2025] Open
Abstract
Oesophageal adenocarcinoma (OAC) is the 7th most common cancer in the United Kingdom (UK) and remains a significant health challenge. This study presents a proteomic analysis of seven OAC donors complementing our previous neoantigen identification study of their human leukocyte antigen (HLA) immunopeptidomes. Our small UK cohort were selected from donors undergoing treatment for OAC. We used label-free mass spectrometry proteomics to compare OAC tumour tissue to matched normal adjacent tissue (NAT) to quantify expression of 3552 proteins. We identified differential expression of a number of proteins previously linked to OAC and other cancers including common markers of tumourigenesis and immunohistological markers, as well as enrichment of processes and pathways relating to RNA processing and the immune system. Our findings also offer insight into the role of the protein stability in the generation of an OAC neoantigen we previously identified. These results provide independent corroboration of existing oesophageal adenocarcinoma biomarker studies that may inform future diagnostic and therapeutic research.
Collapse
Affiliation(s)
- Ben Nicholas
- Centre for Proteomic Research, Biological Sciences and Institute for Life Sciences, Building 85, University of Southampton, Southampton, United Kingdom,
- Centre for Cancer Immunology and Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Alistair Bailey
- Centre for Proteomic Research, Biological Sciences and Institute for Life Sciences, Building 85, University of Southampton, Southampton, United Kingdom,
- Centre for Cancer Immunology and Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Katy J. McCann
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Robert C. Walker
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Peter Johnson
- Cancer Research UK Clinical Centre, University of Southampton, Southampton, United Kingdom,
| | - Tim Elliott
- Centre for Cancer Immunology and Institute for Life Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
- Oxford Cancer Centre for Immuno-Oncology and CAMS-Oxford Institute, Nuffield Department of Medicine, University of Oxford, Southampton, United Kingdom
| | - Tim J. Underwood
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom,
| | - Paul Skipp
- Centre for Proteomic Research, Biological Sciences and Institute for Life Sciences, Building 85, University of Southampton, Southampton, United Kingdom,
| |
Collapse
|
67
|
Knott SJ, Tucholski T, Josyer H, Inman D, Friedl A, Zhu Y, Ge Y, Ponik SM. Deciphering Proteoform Landscape of Mammary Carcinoma by Top-Down Proteomics. J Proteome Res 2025; 24:1425-1438. [PMID: 39936522 PMCID: PMC12006981 DOI: 10.1021/acs.jproteome.4c01044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Defining the proteoform landscape of breast cancer can provide unique insights into the signaling pathways driving disease progression. While bottom-up proteomics has been utilized to profile breast cancer, it lacks the ability to capture intact proteoforms that may underpin the disease. Top-down proteomics is ideally suited to characterize intact proteoforms; however, most top-down proteomics studies have been limited to low molecular weight (MW) proteins (<50 kDa). Herein, we employed a two-dimensional (2D) liquid chromatography combining size exclusion chromatography (SEC) with reverse phase chromatography (RPC) followed by high-resolution mass spectrometry (MS) to expand the coverage for high MW proteoforms. Using this 2D-SEC-RPC-MS approach, we observed a 5-fold increase in the detection of high MW proteoforms (>50 kDa) compared to the conventional 1D-RPC-MS. SEC separation significantly enhanced the detection of high MW proteoforms (>104 kDa), including intermediate filament proteins, vimentin and keratins. Based on accurate mass measurements and MS/MS data, we identified 775 proteoforms from both TFA and HEPES extracts and detected PTMs, such as acetylation, glutathionylation, and myristoylation. Pathway analysis uncovered many proteoforms involved in processes dysregulated in cancer progression. Overall, our findings illustrate the power of top-down proteomics in defining the proteoform landscape of breast carcinoma.
Collapse
Affiliation(s)
- Samantha J. Knott
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave., Madison, Wisconsin 53706, USA
| | - Trisha Tucholski
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave., Madison, Wisconsin 53706, USA
| | - Harini Josyer
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin 53705, USA
| | - David Inman
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin 53705, USA
| | - Andreas Friedl
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1685 Highland Ave., Madison, Wisconsin 53705, USA
| | - Yanlong Zhu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin 53705, USA
- Human Proteomics Program, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin, 53705, USA
| | - Ying Ge
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Ave., Madison, Wisconsin 53706, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin 53705, USA
- Human Proteomics Program, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin, 53705, USA
| | - Suzanne M. Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin 53705, USA
- Carbone Cancer Center, University of Wisconsin-Madison, 1111 Highland Ave., Madison, Wisconsin, 53705, USA
| |
Collapse
|
68
|
Gabiatti BP, Freire ER, Odenwald J, de Freitas Nascimento J, Holetz F, Carrington M, Kramer S, Zoltner M. Trypanosomes lack a canonical EJC but possess an UPF1 dependent NMD-like pathway. PLoS One 2025; 20:e0315659. [PMID: 40053537 PMCID: PMC11888146 DOI: 10.1371/journal.pone.0315659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 11/28/2024] [Indexed: 03/09/2025] Open
Abstract
The exon junction complex (EJC) is a key player in metazoan mRNA quality control and is placed upstream of the exon-exon junction after splicing. Its inner core is composed of Magoh, Y14, eIF4AIII and BTZ and the outer core of proteins involved in mRNA splicing (CWC22), export (Yra1), translation (PYM) and nonsense mediated decay (NMD, UPF1/2/3). Trypanosoma brucei encodes only two genes with introns, but all mRNAs are processed by trans-splicing. The presence of three core EJC proteins and a potential BTZ homologue (Rbp25) in trypanosomes has been suggested to adapt of the EJC function to mark trans-spliced mRNAs. We analysed trypanosome EJC components and noticed major differences between eIF4AIII and Magoh/Y14: (i) whilst eIF4AIII is essential, knocking out both Magoh and Y14 elicits only a mild growth phenotype (ii) eIF4AIII localization is mostly nucleolar, while Magoh and Y14 are nucleolar and nucleoplasmic but excluded from the cytoplasm (iii) eIF4AIII associates with nucleolar proteins and the splicing factor CWC22, but not with Y14 or Magoh, while Magoh and Y14 associate with each other, but not with eIF4AIII, CWC22 or nucleolar proteins. Our data argue against the presence of a functional EJC in trypanosomes, but indicate that eIF4AIII adopted non-EJC related, essential functions, while Magoh and Y14 became redundant. Trypanosomes also possess homologues to the NMD proteins UPF1 and UPF2. Depletion of UPF1 causes only a minor reduction in growth and phylogenetic analyses show several independent losses of UPF1 and UPF2, as well as complete loss of UPF3 in the Kinetoplastida group, indicating that UPF1-dependent NMD is not essential. Regardless, we demonstrate that UPF1 depletion restores the mRNA levels of a PTC reporter. Altogether, we show that the almost intron-less trypanosomes are in the process of losing the canonical EJC/NMD pathways: Y14 and Magoh have become redundant and the still-functional UPF1-dependent NMD pathway is not essential.
Collapse
Affiliation(s)
| | | | - Johanna Odenwald
- Department of Cell and Developmental Biology, University of Würzburg, Würzburg, Germany
| | | | - Fabiola Holetz
- Carlos Chagas Institute (ICC), FIOCRUZ/PR, Curitiba, Brazil
| | - Mark Carrington
- Department of Biochemistry, Cambridge University, Cambridge, United Kingdom
| | - Susanne Kramer
- Department of Cell and Developmental Biology, University of Würzburg, Würzburg, Germany
| | - Martin Zoltner
- Department of Parasitology, Faculty of Science, Charles University in Prague, Biocev, Vestec, Czech Republic
| |
Collapse
|
69
|
Liu X, Dawson SL, Gygi SP, Paulo JA. Isobaric Tagging and Data Independent Acquisition as Complementary Strategies for Proteome Profiling on an Orbitrap Astral Mass Spectrometer. J Proteome Res 2025; 24:1414-1424. [PMID: 39937051 DOI: 10.1021/acs.jproteome.4c01107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Comprehensive global proteome profiling that is amenable to high throughput processing will broaden our understanding of complex biological systems. Here, we evaluate two leading mass spectrometry techniques, Data Independent Acquisition (DIA) and Tandem Mass Tagging (TMT), for extensive protein abundance profiling. DIA provides label-free quantification with a broad dynamic range, while TMT enables multiplexed analysis using isobaric tags for efficient cross-sample comparisons. We analyzed 18 samples, including four cell lines (IHCF, HCT116, HeLa, MCF7) under standard growth conditions, in addition to IHCF treated with two H2O2 concentrations, all in triplicate. Experiments were conducted on an Orbitrap Astral mass spectrometer, employing Field Asymmetric Ion Mobility Spectrometry (FAIMS). Despite utilizing different acquisition strategies, both the DIA and TMT approaches achieved comparable proteome depth and quantitative consistency, with each method quantifying over 10,000 proteins across all samples, with marginally higher protein-level precision for the TMT strategy. Relative abundance correlation analysis showed strong agreement at both peptide and protein levels. Our findings highlight the complementary strengths of DIA and TMT for high-coverage proteomic studies, providing flexibility in method selection based on specific experimental needs.
Collapse
Affiliation(s)
- Xinyue Liu
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Shane L Dawson
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
70
|
Ivanov MV, Kopeykina AS, Kazakova EM, Tarasova IA, Sun Z, Postoenko VI, Yang J, Gorshkov MV. Modified Decision Tree with Custom Splitting Logic Improves Generalization across Multiple Brains' Proteomic Data Sets of Alzheimer's Disease. J Proteome Res 2025; 24:1053-1066. [PMID: 39984290 DOI: 10.1021/acs.jproteome.4c00677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
Many factors negatively affect a generalization of the findings in discovery proteomics. They include differentiation between patient cohorts, a variety of experimental conditions, etc. We presented a machine-learning-based workflow for proteomics data analysis, aiming at improving generalizability across multiple data sets. In particular, we customized the decision tree model by introducing a new parameter, min_groups_leaf, which regulates the presence of the samples from each data set inside the model's leaves. Further, we analyzed a trend for the feature importance's curve as a function of the novel parameter for feature selection to a list of proteins with significantly improved generalization. The developed workflow was tested using five proteomic data sets obtained for post-mortem human brain samples of Alzheimer's disease. The data sets consisted of 535 LC-MS/MS acquisition files. The results were obtained for two different pipelines of data processing: (1) MS1-only processing based on DirectMS1 search engine and (2) a standard MS/MS-based one. Using the developed workflow, we found seven proteins with expression patterns that were unique for asymptomatic Alzheimer patients. Two of them, Serotransferrin TRFE and DNA repair nuclease APEX1, may be potentially important for explaining the lack of dementia in patients with the presence of neuritic plaques and neurofibrillary tangles.
Collapse
Affiliation(s)
- Mark V Ivanov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Anna S Kopeykina
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Elizaveta M Kazakova
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Irina A Tarasova
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Zhao Sun
- Clinical Systems Biology Key Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou 450052, China
| | - Valeriy I Postoenko
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| | - Jinghua Yang
- Clinical Systems Biology Key Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Institute of Infection and Immunity, Henan Academy of Innovations in Medical Science, Zhengzhou 450052, China
| | - Mikhail V Gorshkov
- V. L. Talrose Institute for Energy Problems of Chemical Physics, N. N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119334, Russia
| |
Collapse
|
71
|
Martinez-Seidel F, Suwanchaikasem P, Gentry-Torfer D, Rajarathinam Y, Ebert A, Erban A, Firmino A, Nie S, Leeming M, Williamson N, Roessner U, Kopka J, Boughton BA. Remodelled ribosomal populations synthesize a specific proteome in proliferating plant tissue during cold. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230384. [PMID: 40045790 PMCID: PMC11883437 DOI: 10.1098/rstb.2023.0384] [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: 12/15/2023] [Revised: 06/21/2024] [Accepted: 09/11/2024] [Indexed: 03/09/2025] Open
Abstract
Plant acclimation occurs through system-wide mechanisms that include proteome shifts, some of which occur at the level of protein synthesis. All proteins are synthesized by ribosomes. Rather than being monolithic, transcript-to-protein translation machines, ribosomes can be selective and cause proteome shifts. In this study, we use apical root meristems of germinating seedlings of the monocotyledonous plant barley as a model to examine changes in protein abundance and synthesis during cold acclimation. We measured metabolic and physiological parameters that allowed us to compare protein synthesis in the cold to optimal rearing temperatures. We demonstrated that the synthesis and assembly of ribosomal proteins are independent processes in root proliferative tissue. We report the synthesis and accumulation of various macromolecular complexes and propose how ribosome compositional shifts may be associated with functional proteome changes that are part of successful cold acclimation. Our study indicates that translation initiation is limiting during cold acclimation while the ribosome population is remodelled. The distribution of the triggered ribosomal protein heterogeneity suggests that altered compositions may confer 60S subunits selective association capabilities towards translation initiation complexes. To what extent selective translation depends on heterogeneous ribo-proteome compositions in barley proliferative root tissue remains a yet unresolved question.This article is part of the discussion meeting issue 'Ribosome diversity and its impact on protein synthesis, development and disease'.
Collapse
Affiliation(s)
- Federico Martinez-Seidel
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, CO, USA
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Pipob Suwanchaikasem
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Dione Gentry-Torfer
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Yogeswari Rajarathinam
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Alina Ebert
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Alexander Erban
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Alexandre Firmino
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Shuai Nie
- Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Leeming
- Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria, Australia
- School of Chemistry, The University of Melbourne, Parkville, Victoria, Australia
| | - Nicholas Williamson
- Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ute Roessner
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
- Research School of Biology, The Australian National University, Acton, Australia
| | - Joachim Kopka
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Berin A. Boughton
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
- La Trobe Institute of Sustainable Agriculture and Food, La Trobe University, Bundoora, Victoria3083, Australia
| |
Collapse
|
72
|
Babu M, Rao RM, Babu A, Jerom JP, Gogoi A, Singh N, Seshadri M, Ray A, Shelley BP, Datta A. Antioxidant Effect of Naringin Demonstrated Through a Bayes' Theorem Driven Multidisciplinary Approach Reveals its Prophylactic Potential as a Dietary Supplement for Ischemic Stroke. Mol Neurobiol 2025; 62:3918-3933. [PMID: 39352635 DOI: 10.1007/s12035-024-04525-6] [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: 01/31/2024] [Accepted: 09/23/2024] [Indexed: 01/03/2025]
Abstract
Naringin (NAR), a flavanone glycoside, occurs widely in citrus fruits, vegetables, and alcoholic beverages. Despite evidence of the neuroprotective effects of NAR on animal models of ischemic stroke, brain cell-type-specific data about the antioxidant efficacy of NAR and possible protein targets of such beneficial effects are limited. Here, we demonstrate the brain cell type-specific prophylactic role of NAR, an FDA-listed food additive, in an in vitro oxygen-glucose deprivation (OGD) model of cerebral ischemia using MTT and DCFDA assays. Using Bayes' theorem-based predictive model, we first ranked the top-10 protein targets (ALDH2, ACAT1, CTSB, FASN, LDHA, PTGS1, CTSD, LGALS1, TARDBP, and CDK1) from a curated list of 289 NAR-interacting proteins in neurons that might be mediating its antioxidant effect in the OGD model. When preincubated with NAR for 2 days, N2a and CTX-TNA2 cells could withstand up to 8 h of OGD without a noticeable decrease in cell viability. This cerebroprotective effect is partly mediated by reducing intracellular ROS production in the above two brain cell types. The antioxidant effect of NAR was comparable with the equimolar (50 µM) concentration of clinically used ROS-scavenger and neuroprotective edaravone. Molecular docking of NAR with the top-10 protein targets from Bayes' analysis showed the lowest binding energy for CDK1 (- 8.8 kcal/M). Molecular dynamics simulation analysis showed that NAR acts by inhibiting CDK1 by stably occupying its ATP-binding cavity. Considering diet has been listed as a risk factor for stroke, NAR may be explored as a component of functional food for stroke or related neurological disorders.
Collapse
Affiliation(s)
- Manju Babu
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Rajas M Rao
- Division of Data Analytics, Bioinformatics and Structural Biology, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Anju Babu
- Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal, 462066, MP, India
| | | | - Anaekshi Gogoi
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Nikhil Singh
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Meenakshi Seshadri
- Department of Pharmacology, Yenepoya Pharmacy College and Research Center, Naringana, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Animikh Ray
- Father Muller Research Center, Father Muller Medical College, Mangalore, 575002, Karnataka, India
| | - Bhaskara P Shelley
- Department of Neurology, Yenepoya Medical College, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India
| | - Arnab Datta
- Laboratory of Translational Neuroscience, Division of Neuroscience, Yenepoya Research Center, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, 575018, Karnataka, India.
- Department of Pharmacology, Yenepoya Pharmacy College and Research Center, Naringana, Deralakatte, Mangalore, 575018, Karnataka, India.
| |
Collapse
|
73
|
Lust B, Matthews JL, Oakley CA, Lewis RE, Mendis H, Peng L, Grossman AR, Weis VM, Davy SK. The Influence of Symbiont Identity on the Proteomic and Metabolomic Responses of the Model Cnidarian Aiptasia to Thermal Stress. Environ Microbiol 2025; 27:e70073. [PMID: 40056008 PMCID: PMC11889536 DOI: 10.1111/1462-2920.70073] [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: 10/10/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 05/13/2025]
Abstract
We examined the effects of symbiont identity and heat stress on the host metabolome and proteome in the cnidarian-dinoflagellate symbiosis. Exaiptasia diaphana ('Aiptasia') was inoculated with its homologous (i.e., native) symbiont Breviolum minutum or a heterologous (i.e., non-native) symbiont (Symbiodinium microadriaticum; Durusdinium trenchii) and thermally stressed. Integrated metabolome and proteome analyses characterised host thermal responses between symbioses, with clear evidence of enhanced nutritional deprivation and cellular stress in hosts harbouring heterologous symbionts following temperature stress. Host metabolomes were partially distinct at the control temperature; however, thermal stress caused metabolomes of anemones containing the two heterologous symbionts to become more alike and more distinct from those containing B. minutum. While these patterns could be partly explained by innate symbiont-specific differences, they may also reflect differences in symbiont density, as under control conditions D. trenchii attained 60% and S. microadriaticum 15% of the density attained by B. minutum, and at elevated temperature only D. trenchii-colonised anemones bleached (60% loss). Our findings add to a growing literature that highlights the physiological limits of partner switching as a means of adaptation to global warming. However, we also provide tentative evidence for improved metabolic functioning with a heterologous symbiont (D. trenchii) after sustained symbiosis.
Collapse
Affiliation(s)
- Bobby Lust
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
| | - Jennifer L. Matthews
- Climate Change ClusterUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Clinton A. Oakley
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
| | - Robert E. Lewis
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
| | - Himasha Mendis
- Metabolomics Australia, School of BotanyThe University of MelbourneParkvilleVictoriaAustralia
| | - Lifeng Peng
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
| | - Arthur R. Grossman
- Department of Biosphere Science and EngineeringThe Carnegie Institution for Science, Stanford UniversityStanfordCaliforniaUSA
| | - Virginia M. Weis
- Department of Integrative BiologyOregon State UniversityCorvallisOregonUSA
| | - Simon K. Davy
- School of Biological SciencesVictoria University of WellingtonWellingtonNew Zealand
| |
Collapse
|
74
|
Shi SM, Suh RJ, Shon DJ, Garcia FJ, Buff JK, Atkins M, Li L, Lu N, Sun B, Luo J, To NS, Cheung TH, McNerney MW, Heiman M, Bertozzi CR, Wyss-Coray T. Glycocalyx dysregulation impairs blood-brain barrier in ageing and disease. Nature 2025; 639:985-994. [PMID: 40011765 PMCID: PMC11946907 DOI: 10.1038/s41586-025-08589-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/03/2025] [Indexed: 02/28/2025]
Abstract
The blood-brain barrier (BBB) is highly specialized to protect the brain from harmful circulating factors in the blood and maintain brain homeostasis1,2. The brain endothelial glycocalyx layer, a carbohydrate-rich meshwork composed primarily of proteoglycans, glycoproteins and glycolipids that coats the BBB lumen, is a key structural component of the BBB3,4. This layer forms the first interface between the blood and brain vasculature, yet little is known about its composition and roles in supporting BBB function in homeostatic and diseased states. Here we find that the brain endothelial glycocalyx is highly dysregulated during ageing and neurodegenerative disease. We identify significant perturbation in an underexplored class of densely O-glycosylated proteins known as mucin-domain glycoproteins. We demonstrate that ageing- and disease-associated aberrations in brain endothelial mucin-domain glycoproteins lead to dysregulated BBB function and, in severe cases, brain haemorrhaging in mice. Finally, we demonstrate that we can improve BBB function and reduce neuroinflammation and cognitive deficits in aged mice by restoring core 1 mucin-type O-glycans to the brain endothelium using adeno-associated viruses. Cumulatively, our findings provide a detailed compositional and structural mapping of the ageing brain endothelial glycocalyx layer and reveal important consequences of ageing- and disease-associated glycocalyx dysregulation on BBB integrity and brain health.
Collapse
Affiliation(s)
- Sophia M Shi
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ryan J Suh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - D Judy Shon
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Francisco J Garcia
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Josephine K Buff
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Micaiah Atkins
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
| | - Lulin Li
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Nannan Lu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Bryan Sun
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Ning-Sum To
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Tom H Cheung
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Daniel and Mayce Yu Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - M Windy McNerney
- Department of Psychiatry, Stanford University School of Medicine, Stanford, CA, USA
- MIRECC, Department of Veterans Affairs, Palo Alto, CA, USA
| | - Myriam Heiman
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| | - Tony Wyss-Coray
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA.
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA.
| |
Collapse
|
75
|
Demoures B, Soulet F, Descarpentrie J, Galeano-Otero I, Sanchez Collado J, Casado M, Smani T, González A, Alves I, Lalloué F, Masri B, Rascol E, Dupuy JW, Dourthe C, Saltel F, Raymond AA, Badiola I, Evrard S, Villoutreix B, Pernot S, Siegfried G, Khatib AM. Repression of apelin Furin cleavage sites provides antimetastatic strategy in colorectal cancer. EMBO Mol Med 2025; 17:504-534. [PMID: 39962271 PMCID: PMC11904221 DOI: 10.1038/s44321-025-00196-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
The adipokine apelin has been directly implicated in various physiological processes during embryogenesis and human cancers. Nevertheless, the importance of the conversion of its precursor proapelin to mature apelin in tumorigenesis remains unknown. In this study, we identify Furin as the cellular proprotein convertase responsible for proapelin cleavage. We explore the therapeutic potential of targeting proapelin cleavage sites in metastatic colorectal cancer by introducing apelin-dm, a modified variant resulting from alteration in proapelin cleavage sites. Apelin-dm demonstrates efficacy in inhibiting tumor growth, promoting cell death, suppressing angiogenesis, and early colorectal liver metastasis events. Proteomic analysis reveals reciprocal regulation between apelin and apelin-dm on proteins associated with clinical outcomes in colon cancer patients. Apelin-dm emerges as a modulator of apelin receptor dynamics, influencing affinity, internalization, and repression of apelin signaling linked to various protein kinases. Pharmacokinetic and toxicity assessments confirm the specificity, safety, and stability of apelin-dm, as well as its facile hepatic metabolism. These findings position targeting proapelin cleavage as a promising therapeutic strategy against metastatic colorectal cancer, paving the way for further clinical exploration.
Collapse
Affiliation(s)
- Béatrice Demoures
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
| | - Fabienne Soulet
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
| | - Jean Descarpentrie
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
| | - Isabel Galeano-Otero
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
| | - José Sanchez Collado
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
| | - Maria Casado
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
- Department of Cell Biology and Histology, University of the Basque Country, B° Sarriena sn, 48940, Leioa, Spain
| | - Tarik Smani
- Institute of Biomedicine of Seville, University Hospital of Virgen del Rocío/University of Seville/CSIC, Avenida Manuel Siurot s/n, 41013, Seville, Spain
| | - Alvaro González
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
| | - Isabel Alves
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, Bordeaux, France
| | - Fabrice Lalloué
- EA3842- CAPTuR, GEIST, Faculté de Médecine, Université de Limoges, 2 rue du Dr Marcland, 87025 Cedex, Limoges, France
| | - Bernard Masri
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, 75014, Paris, France
| | - Estelle Rascol
- Univ. Bordeaux, CNRS, Bordeaux INP, CBMN, Bordeaux, France
| | - Jean-William Dupuy
- Bordeaux Protéome, F-33000, Bordeaux, France
- Oncoprot Platform, TBM-Core US 005, Bordeaux, France
| | - Cyril Dourthe
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
- Oncoprot Platform, TBM-Core US 005, Bordeaux, France
| | - Frédéric Saltel
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
- Oncoprot Platform, TBM-Core US 005, Bordeaux, France
| | - Anne-Aurélie Raymond
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
- Oncoprot Platform, TBM-Core US 005, Bordeaux, France
| | - Iker Badiola
- Department of Cell Biology and Histology, University of the Basque Country, B° Sarriena sn, 48940, Leioa, Spain
| | - Serge Evrard
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
- Institut Bergonié, Bordeaux, France
| | - Bruno Villoutreix
- Université de Paris, Inserm UMR 1141, Robert-Debré Hospital, 75019, Paris, France
| | - Simon Pernot
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France
- Institut Bergonié, Bordeaux, France
| | - Géraldine Siegfried
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France.
| | - Abdel-Majid Khatib
- University of Bordeaux, Bordeaux Institute of Oncology (BRIC)-UMR1312, Bordeaux, France.
- Institut Bergonié, Bordeaux, France.
| |
Collapse
|
76
|
Dellar ER, Vendrell I, Amein B, Lester DG, Edmond EC, Yoganathan K, Dharmadasa T, Sogorb‐Esteve A, Fischer R, Talbot K, Rohrer JD, Turner MR, Thompson AG. Elevated Cerebrospinal Fluid Ubiquitin Carboxyl-Terminal Hydrolase Isozyme L1 in Asymptomatic C9orf72 Hexanucleotide Repeat Expansion Carriers. Ann Neurol 2025; 97:449-459. [PMID: 39548852 PMCID: PMC11831881 DOI: 10.1002/ana.27133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/29/2024] [Accepted: 10/25/2024] [Indexed: 11/18/2024]
Abstract
OBJECTIVE To identify biochemical changes in individuals at higher risk of developing amyotrophic lateral sclerosis (ALS) or frontotemporal dementia (FTD) via C9orf72 hexanucleotide repeat expansion (HRE) heterozygosity. METHODS Cross-sectional observational study of 48 asymptomatic C9orf72 HRE carriers, 39 asymptomatic non-carrier controls, 19 people with sporadic ALS, 10 with C9orf72 ALS, 14 with sporadic FTD, and 10 with C9orf72 FTD. Relative abundance of 30 pre-defined cerebrospinal fluid biomarkers of ALS and FTD were compared in asymptomatic C9orf72 HRE carriers and age-matched non-carrier controls. Differential abundance of these proteins was quantified using data independent acquisition mass spectrometry or electro chemiluminescent assay for neurofilament light chain. Unbiased analysis of the entire cerebrospinal fluid proteome was then carried out. RESULTS Ubiquitin carboxyl-hydrolase isozyme L1 levels were higher in asymptomatic C9orf72 HRE carriers compared with age-matched non-carriers (log2fold change 0.20, FDR-adjusted p-value = 0.034), whereas neurofilament light chain levels did not significantly differ. Ubiquitin carboxyl-hydrolase isozyme L1 levels remained elevated after matching of groups by neurofilament levels (p = 0.011), and after adjusting for age, sex, and neurofilament levels. A significant difference was also observed when restricting analysis to younger participants (<37) matched by neurofilament level (p = 0.007). INTERPRETATION Elevated cerebrospinal fluid ubiquitin carboxyl-hydrolase isozyme L1 levels in C9orf72 HRE carriers can occur in the absence of increased neurofilament levels, potentially reflecting either compensatory or pathogenic mechanisms preceding rapid neuronal loss. This brings forward the window on changes associated with the C9orf72 HRE carrier state, with potential to inform understanding of penetrance and approaches to prevention. ANN NEUROL 2025;97:449-459.
Collapse
Affiliation(s)
| | - Iolanda Vendrell
- Target Discovery InstituteCentre for Medicines Discovery, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- Chinese Academy of Medical Science Oxford InstituteUniversity of OxfordOxfordUK
| | - Benazir Amein
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - David G. Lester
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Evan C. Edmond
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Katie Yoganathan
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | - Thanuja Dharmadasa
- The Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkville, VictoriaAustralia
| | - Aitana Sogorb‐Esteve
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
- UK Dementia Research Institute at University College LondonLondonUK
| | - Roman Fischer
- Target Discovery InstituteCentre for Medicines Discovery, Nuffield Department of MedicineUniversity of OxfordOxfordUK
- Chinese Academy of Medical Science Oxford InstituteUniversity of OxfordOxfordUK
| | - Kevin Talbot
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
- Kavli Institute for Nanoscience DiscoveryUniversity of OxfordOxfordUK
| | - Jonathan D. Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease, UCL Queen Square Institute of NeurologyUniversity College LondonLondonUK
| | - Martin R. Turner
- Nuffield Department of Clinical NeurosciencesUniversity of OxfordOxfordUK
| | | |
Collapse
|
77
|
Singh P, Dhir YW, Gupta S, Kaushal A, Kala D, Nagraiik R, Kaushik NK, Noorani MS, Asif AR, Singh B, Aman S, Dhir S. Relevance of proteomics and metabolomics approaches to overview the tumorigenesis and better management of cancer. 3 Biotech 2025; 15:58. [PMID: 39949840 PMCID: PMC11813842 DOI: 10.1007/s13205-025-04222-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Proteomics and metabolomics, integral combination of OMICs platform are gaining prominence in cancer research to enhance scientific knowledge of bio-molecular interactions occurs in the cellular processes during cancer progression. This approach designed to identify potential tools for addressing the complexities of this multifaceted disease. This analysis focussed on the intricate interplay between proteins and metabolites within cancer cells and their surrounding microenvironment. By reviewing current proteomics and metabolomics studies, we aim to gain invaluable insights into tumour biology, progression, and its implication in therapeutic responses. This study highlights the importance of proteomics and metabolomics in discovering therapeutic targets and diagnostic biomarkers for targeted cancer treatment. Proteomics facilitates the analysis of protein expression, modifications and interactions, exemplified by the identification of HER2 mutations leads to development of breast cancer hence targeted therapies like trastuzumab could be initiated. Metabolomics reveals metabolic alternations such as elevated 2-hydroxyglutarate levels in gliomas linked to cancer progression and treatment resistance. The integration of these approaches clarifies complex signalling network driving oncogenesis and paves the way for innovative cancer therapies, including immune cheque point inhibitors. Proteomics and metabolomics have revolutionised cancer biology by revealing intricate signalling networks, metabolic dysregulations, and unique molecular alterations. This information is crucial for early cancer identification and prognosis, and for designing personalized therapeutic strategies. Innovative technologies like artificial intelligence and high-throughput mass spectrometry further enhance the potential of these studies. Fostering multidisciplinary collaboration and data-sharing is essential for maximising the impact of these approaches to cure as well as better management of the cancer.
Collapse
Affiliation(s)
- Pooja Singh
- Department of Bio-sciences & Technology, MMEC, Maharishi Markandeshwar, Deemed to Be University, Mullana, Ambala, Haryana 133207 India
| | - Yashika W. Dhir
- Department of Bio-sciences & Technology, MMEC, Maharishi Markandeshwar, Deemed to Be University, Mullana, Ambala, Haryana 133207 India
| | - Shagun Gupta
- Department of Bio-sciences & Technology, MMEC, Maharishi Markandeshwar, Deemed to Be University, Mullana, Ambala, Haryana 133207 India
| | - Ankur Kaushal
- Department of Bio-sciences & Technology, MMEC, Maharishi Markandeshwar, Deemed to Be University, Mullana, Ambala, Haryana 133207 India
| | - Deepak Kala
- NL-11 Centera Tetrahertz Laboratory, Institute of High-Pressure Physics, Polish Academy of Sciences, 29/37 Sokolowska Street, 01142 Warsaw, Poland
| | - Rupak Nagraiik
- Department of Biotechnology, Graphic Era, Deemed to Be University, Dehradun, Uttarakhand India 248002
| | - Naveen K. Kaushik
- Department of Industrial Biotechnology, College of Biotechnology, Chaudhary Charan Singh Haryana Agricultural University, Hisar, Haryana India
| | - Md Salik Noorani
- Department of Botany, School of Chemical and Life Sciences, Jamia Hamdard, Tughlakabad, New Delhi 110062 India
| | - Abdul R. Asif
- Institute of Clinical Chemistry/UMG Laboratories, University Medical Center Goettingen, Robert Koch-Str.40, 37075 Goettingen, Germany
| | - Bharat Singh
- Department of Bio-sciences & Technology, MMEC, Maharishi Markandeshwar, Deemed to Be University, Mullana, Ambala, Haryana 133207 India
| | - Shahbaz Aman
- Department of Microbiology, MMIMSR, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, Haryana 133207 India
| | - Sunny Dhir
- Department of Bio-sciences & Technology, MMEC, Maharishi Markandeshwar, Deemed to Be University, Mullana, Ambala, Haryana 133207 India
| |
Collapse
|
78
|
Perera TRW, de Ruijter‐Villani M, Gibb Z, Nixon B, Sheridan A, Stout TAE, Swegen A, Skerrett‐Byrne DA. Systemic Changes in Early Pregnancy in the Mare: An Integrated Proteomic Analysis of Blood Plasma, Histotroph, and Yolk Sac Fluid at Day 14 Post-Ovulation. Proteomics Clin Appl 2025; 19:e202400095. [PMID: 39912552 PMCID: PMC11895760 DOI: 10.1002/prca.202400095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/23/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025]
Abstract
PURPOSE Embryo-maternal signaling during the establishment of pregnancy in horses remains one of the biggest mysteries in large animal physiology. Early pregnancy loss represents a major source of economic loss to the breeding industry. This study aimed to investigate the systemic changes associated with early pregnancy by mapping the proteome of blood plasma at 14 days in pregnant and non-pregnant mares. EXPERIMENTAL DESIGN Plasma proteomes were analysed in commercially bred pregnant (n = 17) and non-pregnant (n = 17) Thoroughbred mares at 14 days after ovulation, using high-resolution mass spectrometry. Day 14 histotroph and yolk sac fluid were also profiled and datasets were integrated through pathway analysis. RESULTS We identified 229 total protein IDs, with 12 increased and 10 decreased significantly in pregnant versus non-pregnant plasma. To gain functional insight, these data were aligned with proteomes of 14-day pregnant mare uterine fluid (n = 4; 1358 IDs) and conceptus fluid (soluble proteins within the yolk sac fluid; n = 4; 1152 IDs), and further interrogated using gene ontology databases and pathway analysis. CONCLUSIONS AND CLINICAL RELEVANCE These analyses identified consistent systemic changes in the mare's proteome that indicate a profound and specific immune response to early pregnancy, which appears to precede the systemic endocrine response to pregnancy. Integrated pathway analysis suggests that embryo-maternal interactions in early pregnancy may mimic elements of the virus-host interaction to modulate the maternal immune response. Transthyretin (TTR) and uteroglobin (SCGB1A1) were respectively down- and upregulated in plasma while also present in uterine fluid, and are proposed to be key proteins in early pregnancy establishment. These findings contribute significantly to our knowledge of early pregnancy in the mare and identify potential new avenues for developing clinical approaches to reduce early embryo loss.
Collapse
Affiliation(s)
- Tharangani R. W. Perera
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | | | - Zamira Gibb
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Brett Nixon
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Alecia Sheridan
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - Tom A. E. Stout
- Department of Clinical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Aleona Swegen
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - David A. Skerrett‐Byrne
- Priority Research Centre for Reproductive ScienceSchool of Environmental and Life SciencesCollege of EngineeringScience and EnvironmentThe University of NewcastleCallaghanNew South WalesAustralia
- Infertility and Reproduction Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
- Institute of Experimental GeneticsHelmholtz Zentrum MünchenGerman Research Center for Environmental HealthNeuherbergGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| |
Collapse
|
79
|
Min CW, Gupta R, Lee GH, Cho JH, Kim YJ, Wang Y, Jung KH, Kim ST. Integrative Proteomic and Phosphoproteomic Profiling Reveals the Salt-Responsive Mechanisms in Two Rice Varieties (Oryza Sativa subsp. Japonica and Indica). Proteomics 2025; 25:e202400251. [PMID: 39491529 DOI: 10.1002/pmic.202400251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/09/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024]
Abstract
Salinity stress induces ionic and osmotic imbalances in rice plants that in turn negatively affect the photosynthesis rate, resulting in growth retardation and yield penalty. Efforts have, therefore, been carried out to understand the mechanism of salt tolerance, however, the complexity of biological processes at proteome levels remains a major challenge. Here, we performed a comparative proteome and phosphoproteome profiling of microsome enriched fractions of salt-tolerant (cv. IR73; indica) and salt-susceptible (cv. Dongjin/DJ; japonica) rice varieties. This approach led to the identification of 5856 proteins, of which 473 and 484 proteins showed differential modulation between DJ and IR73 sample sets, respectively. The phosphoproteome analysis led to the identification of a total of 10,873 phosphopeptides of which 2929 and 3049 phosphopeptides showed significant differences in DJ and IR73 sample sets, respectively. The integration of proteome and phosphoproteome data showed activation of ABA and Ca2+ signaling components exclusively in the salt-tolerant variety IR73 in response to salinity stress. Taken together, our results highlight the changes at proteome and phosphoproteome levels and provide a mechanistic understanding of salinity stress tolerance in rice.
Collapse
Affiliation(s)
- Cheol Woo Min
- Department of Plant Bioscience, Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| | - Ravi Gupta
- College of General Education, Kookmin University, Seoul, Republic of Korea
| | - Gi Hyun Lee
- Department of Plant Bioscience, Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| | - Jun-Hyeon Cho
- Sangju Substation, National Institute of Crop Science, Rural Development Administration (RDA), Sangju, Republic of Korea
| | - Yu-Jin Kim
- Department of Life Science and Environmental Biochemistry, Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| | - Yiming Wang
- Key Laboratory of Integrated Management of Crop Disease and Pests, Department of Plant Pathology, Ministry of Education, Nanjing Agricultural University, Nanjing, China
| | - Ki-Hong Jung
- Graduate School of Biotechnology and Crop Biotech Institute, Kyung Hee University, Yongin, Republic of Korea
| | - Sun Tae Kim
- Department of Plant Bioscience, Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| |
Collapse
|
80
|
Bubis JA, Arrey TN, Damoc E, Delanghe B, Slovakova J, Sommer TM, Kagawa H, Pichler P, Rivron N, Mechtler K, Matzinger M. Challenging the Astral mass analyzer to quantify up to 5,300 proteins per single cell at unseen accuracy to uncover cellular heterogeneity. Nat Methods 2025; 22:510-519. [PMID: 39820751 PMCID: PMC11903296 DOI: 10.1038/s41592-024-02559-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 11/06/2024] [Indexed: 01/19/2025]
Abstract
Despite significant advancements in sample preparation, instrumentation and data analysis, single-cell proteomics is currently limited by proteomic depth and quantitative performance. Here we demonstrate highly improved depth of proteome coverage as well as accuracy and precision for quantification of ultra-low input amounts. Using a tailored library, we identify up to 7,400 protein groups from as little as 250 pg of HeLa cell peptides at a throughput of 50 samples per day. Using a two-proteome mix, we check for optimal parameters of quantification and show that fold change differences of 2 can still be successfully determined at single-cell-level inputs. Eventually, we apply our workflow to A549 cells, yielding a proteome coverage ranging from 1,801 to a maximum of >5,300 protein groups from a single cell depending on cell size and search strategy used, which allows for the study of dependencies between cell size and cell cycle phase. Additionally, our workflow enables us to distinguish between in vitro analogs of two human blastocyst lineages: naive human pluripotent stem cells (epiblast) and trophectoderm-like cells. Our data harmoniously align with transcriptomic data, indicating that single-cell proteomics possesses the capability to identify biologically relevant differences within the blastocyst.
Collapse
Affiliation(s)
- Julia A Bubis
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, Vienna, Austria.
| | | | | | | | - Jana Slovakova
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - Theresa M Sommer
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Harunobu Kagawa
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - Peter Pichler
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, Vienna, Austria
| | - Nicolas Rivron
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria
| | - Karl Mechtler
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, Vienna, Austria.
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria.
- Gregor Mendel Institute of Molecular Plant Biology (GMI), Austrian Academy of Sciences, Vienna BioCenter, Vienna, Austria.
| | - Manuel Matzinger
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter, Vienna, Austria.
| |
Collapse
|
81
|
Wang H, Syed AA, Krijgsveld J, Sigismondo G. Isolation of Proteins on Chromatin Reveals Signaling Pathway-Dependent Alterations in the DNA-Bound Proteome. Mol Cell Proteomics 2025; 24:100908. [PMID: 39842777 PMCID: PMC11889358 DOI: 10.1016/j.mcpro.2025.100908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 01/03/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025] Open
Abstract
Signaling pathways often convergence on transcription factors and other DNA-binding proteins that regulate chromatin structure and gene expression, thereby governing a broad range of essential cellular functions. However, the repertoire of DNA-binding proteins is incompletely understood even for the best-characterized pathways. Here, we optimized a strategy for the isolation of Proteins on Chromatin (iPOC) exploiting tagged nucleoside analogs to label the DNA and capture associated proteins, thus enabling the comprehensive, sensitive, and unbiased characterization of the DNA-bound proteome. We then applied iPOC to investigate chromatome changes upon perturbation of the cancer-relevant PI3K-AKT-mTOR pathway. Our results show distinct dynamics of the DNA-bound proteome upon selective inhibition of PI3K, AKT, or mTOR, and we provide evidence how this signaling cascade regulates the DNA-bound status of SUZ12, thereby modulating H3K27me3 levels. Collectively, iPOC is a powerful approach to study the composition of the DNA-bound proteome operating downstream of signaling cascades, thereby both expanding our knowledge of the mechanism of action of the pathway and unveiling novel chromatin modulators that can potentially be targeted pharmacologically.
Collapse
Affiliation(s)
- Huiyu Wang
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Azmal Ali Syed
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeroen Krijgsveld
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty, Heidelberg University, Heidelberg, Germany.
| | - Gianluca Sigismondo
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
82
|
Bourreau C, Navarro E, Cotinat M, Krejbich M, Guillonneau F, Guette C, Boissard A, Henry C, Corre I, Treps L, Clere N. Secretomes From Non-Small Cell Lung Cancer Cells Induce Endothelial Plasticity Through a Partial Endothelial-to-Mesenchymal Transition. Cancer Med 2025; 14:e70707. [PMID: 40028673 PMCID: PMC11873768 DOI: 10.1002/cam4.70707] [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: 08/19/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 03/05/2025] Open
Abstract
AIM The tumor microenvironment (TME) of non-small cell lung cancer (NSCLC) is highly heterogeneous and is involved in tumorigenesis and resistance to therapy. Among the cells of the TME, endothelial cells are associated with the latter processes through endothelial-to-mesenchymal transition (EndMT). During EndMT, endothelial cells (ECs) progressively lose their endothelial phenotype in favor of a mesenchymal phenotype, which favors the production of cancer-associated fibroblasts (CAFs). Our study aimed to investigate the consequences of exposure to different lung tumor secretomes on EC phenotype and plasticity. MATERIALS AND METHODS Conditioned media (CM) were prepared from the tumor cell lines A549, H1755, H23, H1437, and H1975. Proliferation and migration of ECs treated with these CMs were assessed by Cyquant and Incucyte technologies, respectively. The angiogenic capacity of ECs was assessed by following tubulogenesis on Matrigel. Phenotypic changes in treated ECs were detected by flow cytometry. Morphological analysis of actin fibers was performed by immunohistochemistry, while proteomic analysis by mass spectrometry was used to identify the protein content of secretomes. RESULTS A change of the endothelial phenotype was found when human umbilical vein endothelial cells (HUVECs) were treated with different CMs. This phenotypic change was associated with a morphological change, an increase in both stress fiber expression and spontaneous migration. Furthermore, an increase in mesenchymal markers (α-SMA and CD44) confirmed the phenotypic changes. However, the secretomes did not modify the rate of double-labeled cells (vWF+/α-SMA+ or CD31+/CD44+). Proteomic analysis identified potential targets involved in the EndMT with therapeutic relevance. CONCLUSION Taken together, these data suggest that CMs can induce partial EndMT.
Collapse
Affiliation(s)
- Clara Bourreau
- Univ Angers, Inserm, CNRS, MINT, SFR ICATAngersFrance
- Nantes Université, Université d'Angers, CHU Nantes, Inserm, CNRS, CRCI2NANantesFrance
| | - Emilie Navarro
- Nantes Université, Université d'Angers, CHU Nantes, Inserm, CNRS, CRCI2NANantesFrance
| | - Marine Cotinat
- Nantes Université, Université d'Angers, CHU Nantes, Inserm, CNRS, CRCI2NANantesFrance
| | - Morgane Krejbich
- Nantes Université, Université d'Angers, CHU Nantes, Inserm, CNRS, CRCI2NANantesFrance
| | - François Guillonneau
- Nantes Université, Université d'Angers, CHU Nantes, Inserm, CNRS, CRCI2NANantesFrance
- Institut de Cancérologie de l'OuestAngersFrance
| | - Catherine Guette
- Nantes Université, Université d'Angers, CHU Nantes, Inserm, CNRS, CRCI2NANantesFrance
- Institut de Cancérologie de l'OuestAngersFrance
| | | | | | - Isabelle Corre
- Nantes Université, Université d'Angers, CHU Nantes, Inserm, CNRS, CRCI2NANantesFrance
| | - Lucas Treps
- Nantes Université, Université d'Angers, CHU Nantes, Inserm, CNRS, CRCI2NANantesFrance
| | - Nicolas Clere
- Univ Angers, Inserm, CNRS, MINT, SFR ICATAngersFrance
| |
Collapse
|
83
|
Vánská T, Kouřil R, Opatíková M, Ilíková I, Arshad R, Roudnický P, Ilík P. Photosystem II supercomplexes lacking light-harvesting antenna protein LHCB5 and their organization in the thylakoid membrane. PHYSIOLOGIA PLANTARUM 2025; 177:e70167. [PMID: 40128143 PMCID: PMC11932966 DOI: 10.1111/ppl.70167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 03/26/2025]
Abstract
Light-harvesting protein LHCB5 is one of the three minor antenna proteins (LHCB4-6) that connect the core (C) of photosystem II (PSII) with strongly (S) and moderately (M) bound peripheral trimeric antennae (LHCIIs), forming a dimeric PSII supercomplex known as C2S2M2. Plants lacking LHCB4 and LHCB6 do not form C2S2M2, indicating that these minor antenna proteins are crucial for C2S2M2 assembly. However, studies on antisense asLhcb5 plants suggest this may not apply to LHCB5. Using mild clear-native PAGE (CN-PAGE) and electron microscopy (EM), we separated and structurally characterized the C2S2M2 supercomplex from the Arabidopsis lhcb5 mutant. When compared with wild type (WT), the C2S2M2 supercomplexes in the lhcb5 mutant have slightly different positions of S and M trimers and are generally smaller and present in the thylakoid membrane at higher density. Using CN-PAGE, we did not observe any PSII megacomplexes in the lhcb5 mutant, although they are routinely detected by this method in WT. However, we identified the megacomplexes directly in thylakoid membranes via EM, indicating that the megacomplexes are formed but are too labile to be separated. While in WT, both parallel- and non-parallel-associated PSII supercomplexes can be detected in the thylakoid membrane (Nosek et al., 2017, Plant Journal 89, pp. 104-111), only the parallel-associated PSII supercomplexes were found in the lhcb5 mutant. This finding suggests that the formation of non-parallel-associated PSII supercomplexes depends on the presence of LHCB5. The presence of large PSII supercomplexes and megacomplexes, even though less stable, could explain the WT-like photosynthetic characteristics of the lhcb5 mutant.
Collapse
Affiliation(s)
- Tereza Vánská
- Department of BiophysicsFaculty of Science, Palacký UniversityOlomoucCzech Republic
| | - Roman Kouřil
- Department of BiophysicsFaculty of Science, Palacký UniversityOlomoucCzech Republic
| | - Monika Opatíková
- Department of BiophysicsFaculty of Science, Palacký UniversityOlomoucCzech Republic
| | - Iva Ilíková
- Institute of Experimental Botany of the Czech Academy of Sciences, Centre of Plant Structural and Functional GenomicsOlomoucCzech Republic
| | - Rameez Arshad
- Department of BiophysicsFaculty of Science, Palacký UniversityOlomoucCzech Republic
| | - Pavel Roudnický
- Central European Institute of Technology, Masaryk UniversityBrnoCzech Republic
| | - Petr Ilík
- Department of BiophysicsFaculty of Science, Palacký UniversityOlomoucCzech Republic
| |
Collapse
|
84
|
Kapp KL, Garcia-Marques F, Totten SM, Bermudez A, Tanimoto C, Brooks JD, Pitteri SJ. Intact glycopeptide analysis of human prostate tissue reveals site-specific heterogeneity of protein glycosylation in prostate cancer. Glycobiology 2025; 35:cwaf010. [PMID: 40036572 PMCID: PMC11899575 DOI: 10.1093/glycob/cwaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
Approximately 300,000 American men were diagnosed with prostate cancer in 2024. Existing screening approaches based on measuring levels of prostate-specific antigen in the blood lack specificity for prostate cancer. Studying the glycans attached to proteins has the potential to generate new biomarker candidates and/or increase the specificity of existing protein biomarkers, and studying protein glycosylation changes in prostate cancer could also add new information to our understanding of prostate cancer biology. Here, we present the analysis of N-glycoproteins in clinical prostate cancer tissue and patient-matched, non-cancerous adjacent tissue using LC-MS/MS-based intact N-linked glycopeptide analysis. This analysis allowed us to characterize protein N-linked glycosylation changes in prostate cancer at the glycoprotein, glycopeptide, and glycosite levels. Overall, 1894 unique N-glycosites on 7022 unique N-glycopeptides from 1354 unique glycoproteins were identified. Importantly, we observed an overall increase in glycoprotein, glycopeptide, and glycosite counts in prostate cancer tissue than non-cancerous tissue. We identified biological functions enriched in prostate cancer that relate to cancer development. Additionally, we characterized N-glycosite-specific changes in prostate cancer, demonstrating significant meta- and micro-heterogeneity in N-glycan composition in prostate cancer in comparison to non-cancerous tissue. Our findings support the idea that protein glycosylation is heavily impacted and aberrant in prostate cancer and provide examples of N-glycosite-specific changes that could be exploited for more specific markers of prostate cancer.
Collapse
Affiliation(s)
- Kathryn L Kapp
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA 94304, USA
| | - Fernando Garcia-Marques
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA 94304, USA
| | - Sarah M Totten
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA 94304, USA
| | - Abel Bermudez
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA 94304, USA
| | - Cheylene Tanimoto
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA 94304, USA
| | - James D Brooks
- Department of Urology, Stanford University School of Medicine, Center of Academic Medicine, 453 Quarry Rd, Urology-5656, Palo Alto, California 94304, USA
| | - Sharon J Pitteri
- Canary Center at Stanford for Cancer Early Detection, Department of Radiology, Stanford University School of Medicine, 3155 Porter Drive, Palo Alto, CA 94304, USA
| |
Collapse
|
85
|
Bia R, Mitchell G, Javan H, Nickel I, Pierce J, Selzman CH, Franklin S. Proteomic Characterization of Cardioprotective Human Acellular Amniotic Fluid. ACS OMEGA 2025; 10:6918-6926. [PMID: 40028051 PMCID: PMC11865990 DOI: 10.1021/acsomega.4c09451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 03/05/2025]
Abstract
Amniotic fluid-derived products are a promising resource for cell therapy and tissue engineering due to their anti-inflammatory, angiogenic, and antifibrotic properties. Human amniotic fluid (hAF) has been used in medical applications such as wound healing, skin disorders, and ophthalmic conditions. Recently, we demonstrated that hAF is an effective treatment for myocardial ischemia-reperfusion injury in adult rats. However, the protein composition of full-term acellular hAF has remained poorly characterized. To uncover the biologically active components underlying hAF's cardioprotective effects, we conducted a global proteomic analysis of hAF collected from six patients at full-term cesarean sections. Previously shown to improve cardiac function in ischemic rats, these samples were analyzed by using tandem mass spectrometry. We identified 657 proteins, including 148 unique to the deep learning platform Inferys. Bioinformatic analysis revealed that these proteins are involved in immunity, inflammatory responses, cell adhesion, and apoptotic signaling pathways. In addition, these proteins were highly modified, with methylation and deamidation being the most abundant modifications. This study represents the first mass-spectrometry-based characterization of full-term, acellular hAF, suggesting that hAF offers a wide array of immune-modulating proteins working together to provide robust cardioprotection and a valuable treatment for ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ryan Bia
- Nora
Eccles Harrison Cardiovascular Research and Training Institute, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
| | - Grace Mitchell
- Nora
Eccles Harrison Cardiovascular Research and Training Institute, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
- Division
of Cardiothoracic Surgery, University of
Utah School of Medicine, Salt Lake
City, Utah 84112, United States
| | - Hadi Javan
- Nora
Eccles Harrison Cardiovascular Research and Training Institute, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
- Division
of Cardiothoracic Surgery, University of
Utah School of Medicine, Salt Lake
City, Utah 84112, United States
| | - Ian Nickel
- Nora
Eccles Harrison Cardiovascular Research and Training Institute, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
- Division
of Cardiothoracic Surgery, University of
Utah School of Medicine, Salt Lake
City, Utah 84112, United States
| | - Jan Pierce
- Cell
Therapy and Regenerative Medicine Program, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
| | - Craig H. Selzman
- Nora
Eccles Harrison Cardiovascular Research and Training Institute, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
- Division
of Cardiothoracic Surgery, University of
Utah School of Medicine, Salt Lake
City, Utah 84112, United States
| | - Sarah Franklin
- Nora
Eccles Harrison Cardiovascular Research and Training Institute, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
- Department
of Internal Medicine, Cardiology Division, University of Utah School of Medicine, Salt Lake City, Utah 84112, United States
| |
Collapse
|
86
|
Tiwari M, Gas-Pascual E, Goyal M, Popov M, Matsumoto K, Grafe M, Gräf R, Haltiwanger RS, Olszewski N, Orlando R, Samuelson JC, West CM. Novel antibodies detect nucleocytoplasmic O-fucose in protist pathogens, cellular slime molds, and plants. mSphere 2025; 10:e0094524. [PMID: 39912628 PMCID: PMC11853108 DOI: 10.1128/msphere.00945-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 12/18/2024] [Indexed: 02/07/2025] Open
Abstract
Cellular adaptations to change often involve post-translational modifications of nuclear and cytoplasmic proteins. An example found in protists and plants is the modification of serine and threonine residues of dozens to hundreds of nucleocytoplasmic proteins with a single fucose (O-fucose). A nucleocytoplasmic O-fucosyltransferase occurs in the pathogen Toxoplasma gondii, the social amoeba Dictyostelium, and higher plants, where it is called Spy because mutants have a spindly appearance. O-fucosylation, which is required for optimal proliferation of Toxoplasma and Dictyostelium, is paralogous to the O-GlcNAcylation of nucleocytoplasmic proteins of plants and animals that are involved in stress and nutritional responses. O-fucose was first discovered in Toxoplasma using Aleuria aurantia lectin, but its broad specificity for terminal fucose residues on N- and O-linked glycans in the secretory pathway limits its use. Here we present affinity-purified rabbit antisera that are selective for the detection and enrichment of proteins bearing fucose-O-Ser or fucose-O-Thr. These antibodies detect numerous nucleocytoplasmic proteins in Toxoplasma, Dictyostelium, and Arabidopsis, as well as O-fucose occurring on secretory proteins of Dictyostelium and mammalian cells except when blocked by further glycosylation. The antibodies label Toxoplasma, Acanthamoeba, and Dictyostelium in a pattern reminiscent of O-GlcNAc in animal cells including nuclear pores. The O-fucome of Dictyostelium is partially conserved with that of Toxoplasma and is highly induced during starvation-induced development. These antisera demonstrate the unique antigenicity of O-fucose, document the conservation of the O-fucome among unrelated protists, and enable the study of the O-fucomes of other organisms possessing O-fucosyltransferase-like genes.IMPORTANCEO-fucose (O-Fuc), a form of mono-glycosylation on serine and threonine residues of nuclear and cytoplasmic proteins of some parasites, other unicellular eukaryotes, and plants, is understudied because it is difficult to detect owing to its neutral charge and lability during mass spectrometry. Yet, the O-fucosyltransferase enzyme (OFT) is required for optimal growth of the agent for toxoplasmosis, Toxoplasma gondii, and an unrelated protist, the social amoeba Dictyostelium discoideum. Furthermore, O-fucosylation is closely related to the analogous process of O-GlcNAcylation of thousands of proteins of animal cells, where it plays a central role in stress and nutritional responses. O-Fuc is currently best detected using Aleuria aurantia lectin (AAL), but in most organisms, AAL also recognizes a multitude of proteins in the secretory pathway that are modified with fucose in different ways. By establishing the potential to induce highly specific rabbit antisera that discriminate O-Fuc from all other forms of protein fucosylation, this study expands knowledge about the protist O-fucome and opens a gateway to explore the potential occurrence and roles of this intriguing posttranslational modification in bacteria and other protist pathogens such as Acanthamoeba castellanii.
Collapse
Affiliation(s)
- Megna Tiwari
- Center for Tropical and Emerging Global Diseases, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Elisabet Gas-Pascual
- Center for Tropical and Emerging Global Diseases, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, Athens, Georgia, USA
| | - Manish Goyal
- Department of Molecular and Cell Biology, Boston University School of Dental Medicine, Boston, Massachusetts, USA
| | | | | | - Marianne Grafe
- Department of Cell Biology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Ralph Gräf
- Department of Cell Biology, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Robert S. Haltiwanger
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, Athens, Georgia, USA
| | - Neil Olszewski
- Department of Plant & Microbial Biology, University of Minnesota, St. Paul, Minnesota, USA
| | - Ron Orlando
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, Athens, Georgia, USA
- GlycoScientific LLC, Athens, Georgia, USA
| | - John C. Samuelson
- Department of Molecular and Cell Biology, Boston University School of Dental Medicine, Boston, Massachusetts, USA
| | - Christopher M. West
- Center for Tropical and Emerging Global Diseases, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, Athens, Georgia, USA
| |
Collapse
|
87
|
Gao J, Chen H, Yin H, Chen X, Yang Z, Wang Y, Wu J, Tian Y, Shao H, Wen L, Zhou H. Decoding Protein Glycosylation by an Integrative Mass Spectrometry-Based De Novo Sequencing Strategy. JACS AU 2025; 5:702-713. [PMID: 40017757 PMCID: PMC11863158 DOI: 10.1021/jacsau.4c00960] [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: 10/14/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 03/01/2025]
Abstract
Glycoproteins, representing more than 50% of human proteins and most biopharmaceuticals, are crucial for regulating various biological processes. The complexity of multiple glycosylation sites often leads to incomplete sequence coverage and ambiguous glycan modification profiles. Here, we developed an integrative mass spectrometry-based approach for decoding unknown glycoproteins, which is featured with the combination of deglycosylation-mediated de novo sequencing with glycosylation site characterization. We utilized the enzymatic deglycosylation of N-/ O-glycans to achieve comprehensive sequence coverage. Additionally, EThcD fragmentation enables the identification of high-quality long peptides, facilitating precise protein assembly. We subsequently applied this method to de novo sequencing of the highly glycosylated therapeutic fusion protein Etanercept (Enbrel). We also sequenced three new tumor necrosis factor receptor:Fc-fusion biologics with largely unknown sequences, unveiling subtle distinctions in the primary sequences. Furthermore, we characterized N- and O-glycosylation modifications of these proteins at subunit, glycopeptide, and glycan levels. This strategy bridges the gap between the de novo sequencing and glycosylation modification, providing comprehensive information on the primary structure and glycosylation modifications for glycoproteins. Notably, our method could be a robust solution for accurate sequencing of the glycoproteins and has practical value not only in basic research but also in the biopharmaceutical industry.
Collapse
Affiliation(s)
- Jing Gao
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
| | - Hongxu Chen
- School of
Chinese Materia Medica, Nanjing University
of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Hongrui Yin
- NMPA Key
Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai Institute for Food and Drug Control, 1500 Zhangheng Road, Shanghai 201203, China
| | - Xin Chen
- School of
Chinese Materia Medica, Nanjing University
of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Zhicheng Yang
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
- University
of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Yuqiu Wang
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
- Department
of Otolaryngology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Jianhong Wu
- Thermo
Fisher
Scientific, 2517 Jinke
Road, Shanghai 201206, China
| | - Yinping Tian
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
| | - Hong Shao
- NMPA Key
Laboratory for Quality Control of Therapeutic Monoclonal Antibodies, Shanghai Institute for Food and Drug Control, 1500 Zhangheng Road, Shanghai 201203, China
| | - Liuqing Wen
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
- School of
Chinese Materia Medica, Nanjing University
of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
| | - Hu Zhou
- Analytical
Research Center for Organic and Biological Molecules, State Key Laboratory
of Drug Research, Carbohydrate-Based Drug Research Center, Shanghai
Institute of Materia Medica, Chinese Academy
of Sciences, 555 Zuchongzhi
Road, Shanghai 201203, China
- School of
Chinese Materia Medica, Nanjing University
of Chinese Medicine, 138 Xianlin Avenue, Nanjing, Jiangsu 210023, China
- University
of the Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
- School of
Pharmaceutical Science and Technology, Hangzhou Institute for Advanced
Study, University of Chinese Academy of
Sciences, Hangzhou 310024, China
| |
Collapse
|
88
|
Major GS, Herbold CW, Cheng F, Lee A, Zhuang S, Russell AP, Lindsay A. Cardio-metabolic and cytoskeletal proteomic signatures differentiate stress hypersensitivity in dystrophin-deficient mdx mice. J Proteomics 2025; 312:105371. [PMID: 39732163 DOI: 10.1016/j.jprot.2024.105371] [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: 12/19/2024] [Accepted: 12/21/2024] [Indexed: 12/30/2024]
Abstract
Extreme heterogeneity exists in the hypersensitive stress response exhibited by the dystrophin-deficient mdx mouse model of Duchenne muscular dystrophy. Because stress hypersensitivity can impact dystrophic phenotypes, this research aimed to understand the peripheral pathways driving this inter-individual variability. Male and female mdx mice were phenotypically stratified into "stress-resistant" or "stress-sensitive" groups based on their response to two laboratory stressors. Quantitative proteomics of striated muscle revealed that stress-resistant females were most dissimilar from all other groups, with over 250 proteins differentially regulated with stress hypersensitivity. Males showed less proteomic variation with stress hypersensitivity; however, these changes were associated with pathway enrichment. In the heart, stress-sensitive males had significant enrichment of pathways related to mitochondrial ATP synthesis, suggesting that increased cardio-metabolic capacity is associated with stress hypersensitivity in male mdx mice. In both sexes, stress hypersensitivity was associated with greater expression of beta-actin-like protein 2, indicative of altered cytoskeletal organisation. Despite identifying proteomic signatures associated with stress hypersensitivity, these did not correlate with differences in the serum metabolome acutely after a stressor. These data suggest that the heterogeneity in stress hypersensitivity in mdx mice is partially driven by cytoskeletal organisation, but that sex-specific cardio-metabolic reprogramming may also underpin this phenotype. SIGNIFICANCE: Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease which is associated with a premature loss of ambulation and neurocognitive dysfunction. The hypersensitive stress response in DMD is a heterogeneous phenotype which is poorly understood. This study provided the first investigation of the peripheral mechanisms regulating the hypersensitive stress response by undertaking multi-omics analysis of phenotypically stratified mdx mice. Variations in behaviour and the striated muscle proteomic profiles suggest that cardio-metabolic remodelling and cytoskeletal organisation may contribute to this phenotype. This research offers significant insights into understanding how peripheral dystrophin deficiency relates to the cognitive abnormalities seen in patients with DMD.
Collapse
Affiliation(s)
- Gretel S Major
- School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand
| | - Craig W Herbold
- School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand
| | - Flora Cheng
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Albert Lee
- Motor Neuron Disease Research Centre, Macquarie Medical School, Faculty of Medicine, Health, and Human Sciences, Macquarie University, North Ryde, NSW, Australia
| | - Shuzhao Zhuang
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Aaron P Russell
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Angus Lindsay
- School of Biological Sciences, University of Canterbury, Christchurch 8041, New Zealand; Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia; Department of Medicine, University of Otago, Christchurch 8014, New Zealand; Biomolecular Interaction Centre, School of Biological Sciences, University of Canterbury, Christchurch 8140, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland 1010, New Zealand.
| |
Collapse
|
89
|
Anderson JR, Phelan MM, Caamaño-Gutiérrez E, Clegg PD, Rubio-Martinez LM, Peffers MJ. Metabolomic and proteomic stratification of equine osteoarthritis. Equine Vet J 2025. [PMID: 39972657 DOI: 10.1111/evj.14490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/14/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Equine osteoarthritis (OA) is predominantly diagnosed through clinical examination and radiography, leading to detection only after significant joint pathology. The pathogenesis of OA remains unclear and while many medications modify the disease's inflammatory components, no curative or reversal treatments exist. Identifying differentially abundant metabolites and proteins correlated with osteoarthritis severity could improve early diagnosis, track disease progression, and evaluate responses to interventions. OBJECTIVES To identify molecular markers of osteoarthritis severity based on histological and macroscopic grading. STUDY DESIGN Cross-sectional study. METHODS Post-mortem synovial fluid was collected from 58 Thoroughbred racehorse joints and 83 joints from mixed breeds. Joints were histologically and macroscopically scored and categorised by OA and synovitis grade. Synovial fluid nuclear magnetic resonance metabolomic and mass spectrometry proteomic analyses were performed, individually and combined. RESULTS In Thoroughbreds, synovial fluid concentrations of metabolites 2-aminobutyrate, alanine and creatine were elevated for higher OA grades, while glutamate was reduced for both Thoroughbreds and mixed breeds. In mixed breeds, concentrations of three uncharacterised proteins, lipopolysaccharide binding protein and immunoglobulin kappa constant were lower for higher OA grades; concentrations of an uncharacterised protein were higher for OA grade 1 only, and apolipoprotein A1 concentrations were higher for OA grades 1 and 2 compared with lower grades. For Thoroughbreds, gelsolin concentrations were lower for higher OA grades, and afamin was lower at a higher synovitis grade. Correlation analyses of combined metabolomics and proteomics datasets revealed 58 and 32 significant variables for Thoroughbreds and mixed breeds, respectively, with correlations from -0.48 to 0.42 and -0.44 to 0.49. MAIN LIMITATIONS The study's reliance on post-mortem assessments limits correlation with clinical osteoarthritis severity. CONCLUSIONS Following stratification of equine OA severity through histological and macroscopic grading, synovial fluid metabolomic and proteomic profiling identified markers that may support earlier diagnosis and progression tracking. Further research is needed to correlate these markers with clinical osteoarthritis severity.
Collapse
Affiliation(s)
- James R Anderson
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Veterinary Anatomy, Physiology and Pathology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Marie M Phelan
- NMR Metabolomics Facility, Liverpool Shared Research Facilities (LivSRF) & Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| | - Eva Caamaño-Gutiérrez
- Computational Biology Facility, Technology Directorate & Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Liverpool, UK
| | - Peter D Clegg
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Luis M Rubio-Martinez
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Equine Clinical Science, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Neston, UK
- Sussex Equine Hospital, West Sussex, UK
| | - Mandy J Peffers
- Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
90
|
Di Matteo F, Bonrath R, Pravata V, Schmidt H, Ayo Martin AC, Di Giaimo R, Menegaz D, Riesenberg S, de Vrij FMS, Maccarrone G, Holzapfel M, Straub T, Kushner SA, Robertson SP, Eder M, Cappello S. Neuronal hyperactivity in neurons derived from individuals with gray matter heterotopia. Nat Commun 2025; 16:1737. [PMID: 39966398 PMCID: PMC11836124 DOI: 10.1038/s41467-025-56998-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
Periventricular heterotopia (PH), a common form of gray matter heterotopia associated with developmental delay and drug-resistant seizures, poses a challenge in understanding its neurophysiological basis. Human cerebral organoids (hCOs) derived from patients with causative mutations in FAT4 or DCHS1 mimic PH features. However, neuronal activity in these 3D models has not yet been investigated. Here we show that silicon probe recordings reveal exaggerated spontaneous spike activity in FAT4 and DCHS1 hCOs, suggesting functional changes in neuronal networks. Transcriptome and proteome analyses identify changes in neuronal morphology and synaptic function. Furthermore, patch-clamp recordings reveal a decreased spike threshold specifically in DCHS1 neurons, likely due to increased somatic voltage-gated sodium channels. Additional analyses reveal increased morphological complexity of PH neurons and synaptic alterations contributing to hyperactivity, with rescue observed in DCHS1 neurons by wild-type DCHS1 expression. Overall, we provide new comprehensive insights into the cellular changes underlying symptoms of gray matter heterotopia.
Collapse
Affiliation(s)
- Francesco Di Matteo
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Rebecca Bonrath
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Veronica Pravata
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
| | | | - Ane Cristina Ayo Martin
- International Max Planck Research School for Translational Psychiatry (IMPRS-TP), Munich, Germany
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Rossella Di Giaimo
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Biology, University Federico II, Naples, Italy
| | | | | | - Femke M S de Vrij
- Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | | | - Tobias Straub
- Bioinformatics Core, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Steven A Kushner
- Department of Psychiatry, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Stephen P Robertson
- Department of Women's and Children's Health, University of Otago, Dunedin, New Zealand
| | - Matthias Eder
- Max Planck Institute of Psychiatry, Munich, Germany.
| | - Silvia Cappello
- Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU), Munich, Germany.
- Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
91
|
Li F, Zhang T, Syed A, Elbakry A, Holmer N, Nguyen H, Mukkavalli S, Greenberg RA, D'Andrea AD. CHAMP1 complex directs heterochromatin assembly and promotes homology-directed DNA repair. Nat Commun 2025; 16:1714. [PMID: 39962076 PMCID: PMC11832927 DOI: 10.1038/s41467-025-56834-6] [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: 08/02/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
The CHAMP1 complex, a little-known but highly conserved protein complex consisting of CHAMP1, POGZ, and HP1α, is enriched in heterochromatin though its cellular function in these regions of the genome remain unknown. Here we show that the CHAMP complex promotes heterochromatin assembly at multiple chromosomal sites, including centromeres and telomeres, and promotes homology-directed repair (HDR) of DNA double strand breaks (DSBs) in these regions. The CHAMP1 complex is also required for heterochromatin assembly and DSB repair in highly-specialized chromosomal regions, such as the highly-compacted telomeres of ALT (Alternative Lengthening of Telomeres) positive tumor cells. Moreover, the CHAMP1 complex binds and recruits the writer methyltransferase SETDB1 to heterochromatin regions of the genome and is required for efficient DSB repair at these sites. Importantly, peripheral blood lymphocytes from individuals with CHAMP1 syndrome, an inherited neurologic disorder resulting from heterozygous mutations in CHAMP1, also exhibit defective heterochromatin clustering and defective repair of DSBs, suggesting that a defect in DNA repair underlies this syndrome. Taken together, the CHAMP1 complex has a specific role in heterochromatin assembly and the enhancement of HDR in heterochromatin.
Collapse
Affiliation(s)
- Feng Li
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Tianpeng Zhang
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Aleem Syed
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Amira Elbakry
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Noella Holmer
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Huy Nguyen
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sirisha Mukkavalli
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Roger A Greenberg
- Department of Cancer Biology, Penn Center for Genome Integrity, Basser Center for BRCA, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alan D D'Andrea
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
92
|
Lyu MJA, Du H, Yao H, Zhang Z, Chen G, Huang Y, Ni X, Chen F, Zhao YY, Tang Q, Miao F, Wang Y, Zhao Y, Lu H, Fang L, Gao Q, Qi Y, Zhang Q, Zhang J, Yang T, Cui X, Liang C, Lu T, Zhu XG. A dominant role of transcriptional regulation during the evolution of C 4 photosynthesis in Flaveria species. Nat Commun 2025; 16:1643. [PMID: 39952962 PMCID: PMC11828953 DOI: 10.1038/s41467-025-56901-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 02/02/2025] [Indexed: 02/17/2025] Open
Abstract
C4 photosynthesis exemplifies convergent evolution of complex traits. Herein, we construct chromosome-scale genome assemblies and perform multi-omics analysis for five Flaveria species, which represent evolutionary stages from C3 to C4 photosynthesis. Chromosome-scale genome sequence analyses reveal a gradual increase in genome size during the evolution of C4 photosynthesis attributed to the expansion of transposable elements. Systematic annotation of genes encoding C4 enzymes and transporters identify additional copies of three C4 enzyme genes through retrotranspositions in C4 species. C4 genes exhibit elevated mRNA and protein abundances, reduced protein-to-RNA ratios, and comparable translation efficiencies in C4 species, highlighting a critical role of transcriptional regulation in C4 evolution. Furthermore, we observe an increased abundance of ethylene response factor (ERF) transcription factors and cognate cis-regulatory elements associated with C4 genes regulation. Altogether, our study provides valuable genomic resources for the Flaveria genus and sheds lights on evolutionary and regulatory mechanisms underlying C4 photosynthesis.
Collapse
Affiliation(s)
- Ming-Ju Amy Lyu
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China.
| | - Huilong Du
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, China
- School of Life Sciences, Institute of Life Sciences and Green Development, Hebei University, Baoding, China
| | - Hongyan Yao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, 200438, China
| | - Zhiguo Zhang
- Biotechnology Research Institute/National Key Facility for Gene Resources and Gene Improvement, Chinese Academy of Agricultural Sciences, 100081, Beijing, China
| | - Genyun Chen
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Yuhui Huang
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Xiaoxiang Ni
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Faming Chen
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yong-Yao Zhao
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Qiming Tang
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Fenfen Miao
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yanjie Wang
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
- University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yuhui Zhao
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Hongwei Lu
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Lu Fang
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Qiang Gao
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Yiying Qi
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Key Laboratory of Sugarcane Biology and Genetic Breeding, National Engineering Research Center for Sugarcane, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Qing Zhang
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Key Laboratory of Sugarcane Biology and Genetic Breeding, National Engineering Research Center for Sugarcane, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jisen Zhang
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Key Laboratory of Sugarcane Biology and Genetic Breeding, National Engineering Research Center for Sugarcane, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Tao Yang
- China National GeneBank, Shenzhen, 518120, China
| | - Xuean Cui
- Biotechnology Research Institute/National Key Facility for Gene Resources and Gene Improvement, Chinese Academy of Agricultural Sciences, 100081, Beijing, China
| | - Chengzhi Liang
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, China.
| | - Tiegang Lu
- Biotechnology Research Institute/National Key Facility for Gene Resources and Gene Improvement, Chinese Academy of Agricultural Sciences, 100081, Beijing, China.
| | - Xin-Guang Zhu
- State Key Laboratory of Plant Molecular Genetics, Center of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China.
| |
Collapse
|
93
|
Chu F, Lin A. Detecting Human Contaminant Genetically Variant Peptides in Nonhuman Samples. J Proteome Res 2025; 24:579-588. [PMID: 39705712 DOI: 10.1021/acs.jproteome.4c00718] [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: 12/22/2024]
Abstract
During proteomics data analysis, experimental spectra are searched against a user-defined protein database consisting of proteins that are reasonably expected to be present in the sample. Typically, this database contains the proteome of the organism under study concatenated with expected contaminants, such as trypsin and human keratins. However, there are additional contaminants that are not commonly added to the database. In this study, we describe a new set of protein contaminants and provide evidence that they can be detected in mass spectrometry-based proteomics data. Specifically, we provide evidence that human genetically variant peptides (GVPs) can be detected in nonhuman samples. GVPs are peptides that contain single amino acid polymorphisms that result from nonsynonymous single nucleotide polymorphisms in protein-coding regions of DNA. We reanalyzed previously collected nonhuman data-dependent acquisition (DDA) and data-independent acquisition (DIA) data sets and detected between 0 and 135 GVPs per data set. In addition, we show that GVPs are unlikely to originate from nonhuman sources and that a subset of eight GVPs are commonly detected across data sets.
Collapse
Affiliation(s)
- Fanny Chu
- Chemical and Biological Signatures, Pacific Northwest National Laboratory, Seattle, Washington 98109, United States
| | - Andy Lin
- Chemical and Biological Signatures, Pacific Northwest National Laboratory, Seattle, Washington 98109, United States
| |
Collapse
|
94
|
Nicholas B, Bailey A, McCann KJ, Johnson P, Elliott T, Ottensmeier C, Skipp P. Comparative Analysis of Transcriptomic and Proteomic Expression between Two Non-Small Cell Lung Cancer Subtypes. J Proteome Res 2025; 24:729-741. [PMID: 39772544 PMCID: PMC11811994 DOI: 10.1021/acs.jproteome.4c00773] [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: 09/17/2024] [Revised: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 01/11/2025]
Abstract
Non-small cell lung cancer (NSCLC) is frequently diagnosed late and has poor survival. The two predominant subtypes of NSCLC, adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC), are currently differentially diagnosed using immunohistochemical markers; however, they are increasingly recognized as very different cancer types suggestive of potential for new, more targeted therapies. There are extensive efforts to find more precise and noninvasive differential diagnostic tools. Here, we examined these two NSCLC subtypes for differences that may inform treatment and identify potential novel therapeutic pathways. We presented a comparative analysis of transcriptomic and proteomic expression in tumors from a cohort of 22 NSCLC patients: 8 LUSC and 14 LUAD. Comparing NSCLC subtypes, we found differential gene expression related to cell differentiation for LUSC and cellular structure and immune response regulation for LUAD. Differential protein expression between NSCLC subtypes was related to extracellular structure for LUSC and metabolic processes, including glucose metabolism for LUAD. This direct comparison was more informative about subtype-specific pathways than between each subtype and control (nontumor) tissues. Many of our observations between NSCLC subtypes support and inform existing observations and reveal differences that may aid research seeking to identify and validate novel subtype biomarkers or druggable targets.
Collapse
Affiliation(s)
- Ben Nicholas
- Centre
for Proteomic Research, School of Biological Sciences and Institute
for Life Sciences, University of Southampton, Building 85, Southampton SO17 1BJ ,U.K.
- Centre
for Cancer Immunology and Institute for Life Sciences, Faculty of
Medicine, University of Southampton, Southampton SO16 6YD ,U.K.
| | - Alistair Bailey
- Centre
for Proteomic Research, School of Biological Sciences and Institute
for Life Sciences, University of Southampton, Building 85, Southampton SO17 1BJ ,U.K.
- Centre
for Cancer Immunology and Institute for Life Sciences, Faculty of
Medicine, University of Southampton, Southampton SO16 6YD ,U.K.
| | - Katy J. McCann
- School
of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD ,U.K.
| | - Peter Johnson
- Cancer
Research UK Clinical Centre, University
of Southampton, Southampton SO16 6YD ,U.K.
| | - Tim Elliott
- Centre
for Cancer Immunology and Institute for Life Sciences, Faculty of
Medicine, University of Southampton, Southampton SO16 6YD ,U.K.
- Oxford
Cancer Centre for Immuno-Oncology and CAMS-Oxford Institute, Nuffield
Department of Medicine, University of Oxford, Oxford OX3 7LE ,U.K.
| | - Christian Ottensmeier
- School
of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD ,U.K.
- Institute
of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, U.K.
| | - Paul Skipp
- Centre
for Proteomic Research, School of Biological Sciences and Institute
for Life Sciences, University of Southampton, Building 85, Southampton SO17 1BJ ,U.K.
| |
Collapse
|
95
|
Korovesis D, Mérillat C, Derua R, Verhelst SHL. Proteome selectivity profiling of photoaffinity probes derived from imidazopyrazine-kinase inhibitors. Commun Chem 2025; 8:34. [PMID: 39910186 PMCID: PMC11799219 DOI: 10.1038/s42004-025-01436-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
Kinases are attractive drug targets, but the design of highly selective kinase inhibitors remains challenging. Selectivity may be evaluated against a panel of kinases, or - preferred - in a complex proteome. Probes that allow photoaffinity-labeling of their targets can facilitate this process. Here, we report photoaffinity probes based on the imidazopyrazine scaffold, which is found in several kinase inhibitors and drugs or drug candidates. By chemical proteomics experiments, we find a range of off-targets, which vary between the different probes. In silico analysis suggests that differences between probes may be related to the size, spatial arrangement and rigidity of the imidazopyrazine and its substituent at the 1-position.
Collapse
Affiliation(s)
- Dimitris Korovesis
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Christel Mérillat
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven - University of Leuven, Leuven, Belgium
| | - Rita Derua
- Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven - University of Leuven, Leuven, Belgium
- SyBioMa, KU Leuven, Leuven, Belgium
| | - Steven H L Verhelst
- Department of Cellular and Molecular Medicine, Laboratory of Chemical Biology, KU Leuven - University of Leuven, Leuven, Belgium.
| |
Collapse
|
96
|
Moser LM, Heim C, Koschade SE, Wendel P, Bozkurt S, Harenkamp S, Kreyenberg H, Merker M, Münch C, Gradhand E, Vogler M, Ullrich E, Bönig H, Klusmann JH, Bader P, Wels WS, Rettinger E. CAR-CIK vs. CAR-T: benchmarking novel cytokine-induced killer cells as solid tumor immunotherapy in ErbB2+ rhabdomyosarcoma. Front Immunol 2025; 16:1485817. [PMID: 39963129 PMCID: PMC11831232 DOI: 10.3389/fimmu.2025.1485817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Introduction CAR-T cell therapy, though successful in hematologic malignancies, faces challenges in solid tumors due to limitations of autologous T cells. Cytokine-induced killer (CIK) cells can be given safely across allogeneic barriers and constitute alternative effector cells generated from healthy donors. CIK cells are a heterogenous population of predominantly T cells with a mixed natural killer (NK) phenotype and combine non-MHC-restricted cytotoxicity with potent anti-tumor capacity of the adaptive immune system. Here, we characterize and compare efficacy, phenotypic subpopulations and modes of action of CAR-CIK cells and conventional CAR-T cells from same-donor samples in ErbB2+ rhabdomyosarcoma (RMS). Methods To benchmark CAR-CIK against conventional CAR-T cells, effector cells were generated from same-donor samples and lentivirally transduced with a second generation CD28-CD3ζ CAR. Effector subpopulations and their dynamics upon target cell exposure were phenotypically characterized by flow cytometry. Efficacy was assessed in human ErbB2+ RMS cancer cell lines and primary patient samples in vitro and ex vivo using cytotoxicity and spheroid co-incubation assays. Modes of action were assessed by comparing cytokine secretion profiles using bead-based multiplexed flow cytometry and by liquid chromatography mass spectrometry whole cell proteomics. Finally, we used an in vivo model of RMS mimicking minimal metastatic residual disease to compare anti-tumor potency of CAR-CIK vs. CAR-T cells and to assess their target organ infiltration. Results In vitro assays demonstrated superior cytotoxicity of CAR-CIK cells against RMS cell lines and primary tumor samples. Long-term co-incubation with tumor spheroids led to expansion of CAR-CIK cells and enrichment of CD3+CD56+ TNK cells. CAR-CIK cell cytokine signature showed significantly increased secretion of effector molecules like interferon-γ, perforin and granulysin, and lower secretion of Th2 cytokines IL-2, IL-4 and IL-10. Whole cell proteomics showed corresponding upregulation of chemokine signaling and NK-cytotoxicity pathways in CAR-CIK cells. In NSG mice xenografted with ErbB2+ RMS, a single injection of either CAR-effector cells strongly impeded metastatic tumor development and significantly improved survival. Conclusion Our results demonstrate that CAR-CIK cells are at least equipotent to CAR-T cells. Combined with their favorable safety profile and allogeneic applicability, these findings position CAR-CIK cells as promising immune effectors for solid tumors.
Collapse
Affiliation(s)
- Laura M. Moser
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), Frankfurt am Main, Germany
| | - Catrin Heim
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Sebastian E. Koschade
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), Frankfurt am Main, Germany
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Philipp Wendel
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- Experimental Immunology & Cell Therapy, Department of Pediatrics, Goethe University, Frankfurt am Main, Germany
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Darmstadt, Germany
| | - Süleyman Bozkurt
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Sabine Harenkamp
- Department of Cellular Therapeutics/Cell Processing, Institute for Transfusion Medicine and Immunotherapy, Goethe University, Frankfurt am Main, Germany
| | - Hermann Kreyenberg
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Michael Merker
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Christian Münch
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
- Cardio-Pulmonary Institute, Frankfurt am Main, Germany
| | - Elise Gradhand
- Department of Pediatric and Perinatal Pathology, Dr. Senckenberg Institute of Pathology, Goethe-University Frankfurt, Frankfurt am Main, Germany
| | - Meike Vogler
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- Institute for Experimental Pediatric Hematology and Oncology, Goethe University, Frankfurt am Main, Germany
| | - Evelyn Ullrich
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), Frankfurt am Main, Germany
- Experimental Immunology & Cell Therapy, Department of Pediatrics, Goethe University, Frankfurt am Main, Germany
| | - Halvard Bönig
- Department of Cellular Therapeutics/Cell Processing, Institute for Transfusion Medicine and Immunotherapy, Goethe University, Frankfurt am Main, Germany
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Jan-Henning Klusmann
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), Frankfurt am Main, Germany
| | - Peter Bader
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), Frankfurt am Main, Germany
| | - Winfried S. Wels
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Eva Rettinger
- Division for Stem Cell Transplantation and Immunology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- Department of Pediatrics, Goethe University Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), partner site Frankfurt/Mainz, Frankfurt am Main, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, Frankfurt am Main, Germany
- Universitäres Centrum für Tumorerkrankungen (UCT), Frankfurt am Main, Germany
| |
Collapse
|
97
|
Kirchhoff A, Herzner AM, Urban C, Piras A, Düster R, Mahlberg J, Grünewald A, Schlee-Guimarães TM, Ciupka K, Leka P, Bootz RJ, Wallerath C, Hunkler C, de Regt AK, Kümmerer BM, Christensen MH, Schmidt FI, Lee-Kirsch MA, Günther C, Kato H, Bartok E, Hartmann G, Geyer M, Pichlmair A, Schlee M. RNA-binding proteins hnRNPM and ELAVL1 promote type-I interferon induction downstream of the nucleic acid sensors cGAS and RIG-I. EMBO J 2025; 44:824-853. [PMID: 39707025 PMCID: PMC11791083 DOI: 10.1038/s44318-024-00331-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 12/23/2024] Open
Abstract
The cytosolic nucleic acid sensors RIG-I and cGAS induce type-I interferon (IFN)-mediated immune responses to RNA and DNA viruses, respectively. So far no connection between the two cytosolic pathways upstream of IKK-like kinase activation has been investigated. Here, we identify heterogeneous nuclear ribonucleoprotein M (hnRNPM) as a positive regulator of IRF3 phosphorylation and type-I IFN induction downstream of both cGAS and RIG-I. Combining interactome analysis with genome editing, we further uncover the RNA-binding protein ELAV-like protein 1 (ELAVL1; also known as human antigen R, HuR) as an hnRNPM interactor. Depletion of hnRNPM or ELAVL1 impairs type-I IFN induction by herpes simplex virus 1 or Sendai virus. In addition, we show that hnRNPM and ELAVL1 interact with TANK-binding kinase 1, IκB kinase ε, IκB kinase β, and NF-κB p65. Our confocal microscopy experiments demonstrate cytosolic and perinuclear interactions between hnRNPM, ELAVL1, and TBK1. Furthermore, pharmacological inhibition of ELAVL1 strongly reduces cytokine release from type-I interferonopathy patient fibroblasts. The RNA-binding proteins hnRNPM and ELAVL1 are the first non-redundant regulators to bridge the cGAS/STING and RIG-I/MAVS pathways. Overall, our study characterizes the hnRNPM-ELAVL1 complex as a novel system promoting antiviral defense, pointing to a potential therapeutic target to reduce auto-inflammation in patients with type-I interferonopathies.
Collapse
Affiliation(s)
- Alexander Kirchhoff
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.
| | - Anna-Maria Herzner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
- Department of Cancer Immunology and Immune Modulation, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riß, Germany
| | - Christian Urban
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Antonio Piras
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
| | - Robert Düster
- Institute of Structural Biology, University Hospital Bonn, Bonn, Germany
| | - Julia Mahlberg
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Agathe Grünewald
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Thais M Schlee-Guimarães
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Katrin Ciupka
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Petro Leka
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Robert J Bootz
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Christina Wallerath
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Charlotte Hunkler
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Ann Kristin de Regt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Beate M Kümmerer
- Institute of Virology, University Hospital Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, 53127, Bonn, Germany
| | | | - Florian I Schmidt
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Min Ae Lee-Kirsch
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site Leipzig/Dresden, Dresden, Germany
| | - Claudia Günther
- Department of Dermatology, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Hiroki Kato
- Institute of Cardiovascular Immunology, University Hospital Bonn, Bonn, Germany
| | - Eva Bartok
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
- Unit of Experimental Immunology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Gunther Hartmann
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University Hospital Bonn, Bonn, Germany
| | - Andreas Pichlmair
- Institute of Virology, Technical University of Munich, School of Medicine, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, 81675, Munich, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany.
| |
Collapse
|
98
|
Polasky DA, Lu L, Yu F, Li K, Shortreed MR, Smith LM, Nesvizhskii AI. Quantitative proteome-wide O-glycoproteomics analysis with FragPipe. Anal Bioanal Chem 2025; 417:921-930. [PMID: 38877149 PMCID: PMC11648966 DOI: 10.1007/s00216-024-05382-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/16/2024]
Abstract
Identification of O-glycopeptides from tandem mass spectrometry data is complicated by the near complete dissociation of O-glycans from the peptide during collisional activation and by the combinatorial explosion of possible glycoforms when glycans are retained intact in electron-based activation. The recent O-Pair search method provides an elegant solution to these problems, using a collisional activation scan to identify the peptide sequence and total glycan mass, and a follow-up electron-based activation scan to localize the glycosite(s) using a graph-based algorithm in a reduced search space. Our previous O-glycoproteomics methods with MSFragger-Glyco allowed for extremely fast and sensitive identification of O-glycopeptides from collisional activation data but had limited support for site localization of glycans and quantification of glycopeptides. Here, we report an improved pipeline for O-glycoproteomics analysis that provides proteome-wide, site-specific, quantitative results by incorporating the O-Pair method as a module within FragPipe. In addition to improved search speed and sensitivity, we add flexible options for oxonium ion-based filtering of glycans and support for a variety of MS acquisition methods and provide a comparison between all software tools currently capable of O-glycosite localization in proteome-wide searches.
Collapse
Affiliation(s)
- Daniel A Polasky
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| | - Lei Lu
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Pharmaceutical Chemistry, University of San Francisco, San Francisco, CA, USA
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Kai Li
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | | | - Lloyd M Smith
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 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.
| |
Collapse
|
99
|
Weber M, Fuchs D, Pöschel A, Beebe E, Garajova Z, Jarosch A, Kunz L, Wolski W, Opitz L, Guscetti F, Nolff MC, Markkanen E. Transcriptomic and proteomic profiling identifies feline fibrosarcoma as clinically amenable model for aggressive sarcoma subtypes. Neoplasia 2025; 60:101104. [PMID: 39681068 PMCID: PMC11713505 DOI: 10.1016/j.neo.2024.101104] [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: 09/19/2024] [Revised: 12/06/2024] [Accepted: 12/10/2024] [Indexed: 12/18/2024]
Abstract
Fibrosarcomas (FSA) are malignant mesenchymal tumors characterized by low chemo- and radiosensitivity. Development of novel treatment strategies for human adult FSA is hindered by the low incidence and the absence of suitable clinical models. Interestingly, aggressive FSA occur more frequently in domestic cats, hence potentially representing a clinically amenable model to assess novel therapies such as targeted imaging or theranostics. However, a lack of molecular characterization of FSA and adjacent normal tissue (NT) in both species hinders identification of tumor-specific targets and undermines the translational potential of feline FSA. Combining laser-capture microdissection, RNAsequencing and liquid chromatography-tandem mass spectrometry, we perform comprehensive profiling of 30 feline FSA and matched skeletal muscle, adipose and connective tissue. Clear inter-tissue differences allow identification of significantly upregulated and tumor-exclusive features that represent potential targets for diagnostic and therapeutic approaches. While feline FSA are characterized by hyperactive EIF2, TP53 and MYC signaling, immune-related and neuronal pathways emerge as modulators of tumor aggressiveness and immunosuppression. A high degree of molecular similarity with canine and adult FSA allows identification of tumor targets that are conserved across species. Significant enrichment in DNA repair pathways in feline FSA correlate with aggressive clinical behavior in human soft-tissue sarcoma. Finally, we leverage the molecular profiles to identify vulnerabilities, including sensitivity to ATR and PARP inhibition as potential treatment for feline FSA. In conclusion, this detailed landscape provides a rich resource to identify target candidates and therapeutic vulnerabilities within and across species and supports feline FSA as relevant models for the human disease.
Collapse
Affiliation(s)
- Mikiyo Weber
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Daniel Fuchs
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Amiskwia Pöschel
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Erin Beebe
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Zuzana Garajova
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Armin Jarosch
- Institute of Pathology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Laura Kunz
- Functional Genomics Center Zürich, ETH Zürich/University of Zurich, 8057 Zürich, Switzerland
| | - Witold Wolski
- Functional Genomics Center Zürich, ETH Zürich/University of Zurich, 8057 Zürich, Switzerland
| | - Lennart Opitz
- Functional Genomics Center Zürich, ETH Zürich/University of Zurich, 8057 Zürich, Switzerland
| | - Franco Guscetti
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Mirja C Nolff
- Clinic for Small Animal Surgery, Vetsuisse Faculty, University Animal Hospital, University of Zurich, Zurich, Switzerland.
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland.
| |
Collapse
|
100
|
Sanz-Martinez P, Berkane R, Stolz A. Function of CSNK2/CK2 selectively affects the endoplasmic reticulum and the Golgi apparatus in mtor-mediated autophagy induction. Autophagy 2025; 21:480-486. [PMID: 39178915 PMCID: PMC11760280 DOI: 10.1080/15548627.2024.2395725] [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: 02/20/2024] [Revised: 08/03/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024] Open
Abstract
Selective macroautophagy/autophagy of the endoplasmic reticulum, known as reticulophagy/ER-phagy, is essential to maintain ER homeostasis. We recently showed that members of the autophagy receptor family RETREG/FAM134 are regulated by phosphorylation-dependent ubiquitination. In an unbiased screen we had identified several kinases downstream of MTOR with profound impact on reticulophagy flux, including ATR and CSNK2/CK2. Inhibition of CSNK2 by SGC-CK2-1 prevented regulatory ubiquitination of RETREG1/FAM134B and RETREG3/FAM134C upon autophagy activation as well as the formation of high-density RETREG1- and RETREG3-clusters. Here we report on additional resource data of global proteomics upon CSNK2 and ATR inhibition, respectively. Our data suggests that the function of CSNK2 is mainly limited to the ER/reticulophagy and Golgi/Golgiphagy, while ATR inhibition by VE-822 affects the vast majority of organelles/selective autophagy pathways.Abbreviation: ATRi: ATR inhibitor VE-822; CSNK2i: CSNK2 inhibitor SGC-CK2-1; ER: endoplasmic reticulum.
Collapse
Affiliation(s)
- Pablo Sanz-Martinez
- Institute of Biochemistry 2 (IBC2), Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, Frankfurt am Main, Germany
| | - Rayene Berkane
- Institute of Biochemistry 2 (IBC2), Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, Frankfurt am Main, Germany
| | - Alexandra Stolz
- Institute of Biochemistry 2 (IBC2), Goethe University, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University, Frankfurt am Main, Germany
| |
Collapse
|