1
|
Famà V, Coscujuela Tarrero L, Albanese R, Calviello L, Biffo S, Pelizzola M, Furlan M. Coupling mechanisms coordinating mRNA translation with stages of the mRNA lifecycle. RNA Biol 2025; 22:1-12. [PMID: 40116043 PMCID: PMC11934187 DOI: 10.1080/15476286.2025.2483001] [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: 12/29/2024] [Revised: 03/06/2025] [Accepted: 03/13/2025] [Indexed: 03/23/2025] Open
Abstract
Gene expression involves a series of consequential processes, beginning with mRNA synthesis and culminating in translation. Traditionally studied as a linear sequence of events, recent findings challenge this perspective, revealing coupling mechanisms that coordinate key steps of gene expression, even when spatially and temporally distant. In this review, we focus on translation, the final stage of gene expression, and examine its coupling with key stages of mRNA metabolism: synthesis, processing, export, and decay. For each of these processes, we provide an overview of known instances of coupling with translation. Furthermore, we discuss the role of high-throughput technologies in uncovering these intricate interactions on a genome-wide scale. Finally, we highlight key challenges and propose future directions to advance our understanding of how coupling mechanisms orchestrate robust and adaptable gene expression programs.
Collapse
Affiliation(s)
- Valeria Famà
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan, Italy
- Department of Oncology and Emato-Oncology, University of Milan, Milan, Italy
| | | | | | | | - Stefano Biffo
- National Institute of Molecular Genetics, Fondazione Romeo ed Enrica Invernizzi, INGM, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
| | - Mattia Pelizzola
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Mattia Furlan
- Center for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia (IIT), Milan, Italy
| |
Collapse
|
2
|
Zhang F, Celis-Gutierrez J, Zhang L, Mellado V, Gelard L, Panigot S, Mori D, Lu L, Voisinne G, Vilarnau Wolek C, Mello M, Burlet-Schiltz O, Gonzalez de Peredo A, Fiore F, Roncagalli R, Liang Y, Malissen M, Malissen B. A CARMIL2 gain-of-function mutation suffices to trigger most CD28 costimulatory functions in vivo. J Exp Med 2025; 222:e20250339. [PMID: 40402149 PMCID: PMC12097149 DOI: 10.1084/jem.20250339] [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: 02/13/2025] [Revised: 03/30/2025] [Accepted: 04/23/2025] [Indexed: 05/23/2025] Open
Abstract
Naive T cell activation requires both TCR and CD28 signals. The CARMIL2 cytosolic protein enables CD28-dependent activation of the NF-κB transcription factor via its ability to link CD28 to the CARD11 adaptor protein. Here, we developed mice expressing a mutation named Carmil2QE and mimicking a mutation found in human T cell malignancies. Naive T cells from Carmil2QE mice contained preformed CARMIL2QE-CARD11 complexes in numbers comparable to those assembling in wild-type T cells after CD28 engagement. Such ready-made CARMIL2QE-CARD11 complexes also formed in CD28-deficient mice where they unexpectedly induced most of the functions that normally result from CD28 engagement in a manner that remains antigen-dependent. In turn, tumor-specific T cells expressing Carmil2QE do not require CD28 engagement and thereby escape to both PD-1 and CTLA-4 inhibition. In conclusion, we uncovered the overarching role played by CARMIL2-CARD11 signals among those triggered by CD28 and exploited them to induce potent solid tumor-specific T cell responses in the absence of CD28 ligands and immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Fanghui Zhang
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
- School of Medical
Technology, Xinxiang Medical University,
Xinxiang City, China
| | - Javier Celis-Gutierrez
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
| | - Lichen Zhang
- School of Medical
Technology, Xinxiang Medical University,
Xinxiang City, China
| | - Valentin Mellado
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
- Centre
d’Immunophénomique (CIPHE), Aix Marseille Université,
INSERM, CNRS, Marseille, France
| | - Léna Gelard
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
| | - Sophie Panigot
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
- Centre
d’Immunophénomique (CIPHE), Aix Marseille Université,
INSERM, CNRS, Marseille, France
| | - Daiki Mori
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
- Centre
d’Immunophénomique (CIPHE), Aix Marseille Université,
INSERM, CNRS, Marseille, France
| | - Liaoxun Lu
- School of Medical
Technology, Xinxiang Medical University,
Xinxiang City, China
| | - Guillaume Voisinne
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
| | - Carine Vilarnau Wolek
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
- Centre
d’Immunophénomique (CIPHE), Aix Marseille Université,
INSERM, CNRS, Marseille, France
| | - Marielle Mello
- Centre
d’Immunophénomique (CIPHE), Aix Marseille Université,
INSERM, CNRS, Marseille, France
| | - Odile Burlet-Schiltz
- Institut de
Pharmacologie et de Biologie Structurale (IPBS), Université de
Toulouse, CNRS, Université Toulouse III - Paul Sabatier
(UPS), Toulouse, France
| | - Anne Gonzalez de Peredo
- Institut de
Pharmacologie et de Biologie Structurale (IPBS), Université de
Toulouse, CNRS, Université Toulouse III - Paul Sabatier
(UPS), Toulouse, France
| | - Frédéric Fiore
- Centre
d’Immunophénomique (CIPHE), Aix Marseille Université,
INSERM, CNRS, Marseille, France
| | - Romain Roncagalli
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
| | - Yinming Liang
- School of Medical
Technology, Xinxiang Medical University,
Xinxiang City, China
- Laboratory of Mouse
Genetics, Institute of Psychiatry and Neuroscience, Xinxiang Medical
University, Xinxiang City, China
| | - Marie Malissen
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
- School of Medical
Technology, Xinxiang Medical University,
Xinxiang City, China
- Centre
d’Immunophénomique (CIPHE), Aix Marseille Université,
INSERM, CNRS, Marseille, France
- Laboratory of
Immunophenomics, School of Medical Technology, Xinxiang Medical
University, Xinxiang City, China
| | - Bernard Malissen
- Centre
d’Immunologie de Marseille-Luminy (CIML), Aix Marseille
Université, Institut national de la santé et de la recherche
médicale (INSERM), Centre national de la recherche scientifique
(CNRS), Marseille, France
- School of Medical
Technology, Xinxiang Medical University,
Xinxiang City, China
- Centre
d’Immunophénomique (CIPHE), Aix Marseille Université,
INSERM, CNRS, Marseille, France
- Laboratory of
Immunophenomics, School of Medical Technology, Xinxiang Medical
University, Xinxiang City, China
| |
Collapse
|
3
|
Monteiro R, Alcantud BS, Piersma S, Hendrickx APA, Maaß S, Becher D, Azeredo J, Bathoorn E, van Dijl JM. Outer membrane vesicles of carbapenem-resistant clinical Acinetobacter baumannii isolates protect both the vesicle-producing bacteria and non-resistant bacteria against carbapenems. Microbiol Res 2025; 297:128175. [PMID: 40239429 DOI: 10.1016/j.micres.2025.128175] [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: 09/01/2024] [Revised: 01/23/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025]
Abstract
Infections caused by carbapenem-resistant Acinetobacter baumannii (A. baumannii; CRAb) are associated with high patient morbidity and mortality. The serious threat for human health imposed by CRAb was recently underscored by identification of close-to-untouchable carbapenem- and tetracycline-resistant isolates. Since outer membrane vesicles (OMVs) of Gram-negative bacteria may contribute to antimicrobial resistance, our present study was aimed at investigating OMVs produced by the first two carbapenem- and tetracycline-resistant A. baumannii isolates in Europe. These isolates, denoted CRAb1 and CRAb2, contain large, nearly identical plasmids that specify multiple resistances. Both isolates produce OMVs that were analyzed by differential light scattering, transmission electron microscopy and proteomics. By comparison with OMVs from the plasmid-free non-carbapenem-resistant A. baumannii isolate Ab1, which is an isogenic ancestor of the CRAb1 isolate, we show that plasmid carriage by the CRAb1 and CRAb2 isolates leads to an increased OMV size that is accompanied by increased diversity of the OMV proteome. Our analyses show that OMVs from CRAb1 and CRAb2 are major reservoirs of proteins involved in antimicrobial resistance, including the plasmid-encoded carbapenemases New Delhi metallo-β-lactamase-1 (NDM-1), and carbapenem-hydrolyzing oxacillinase OXA-97 (OXA-97). Here we report that these OMV-borne carbapenemases hydrolyze imipenem and protect otherwise carbapenem-sensitive A. baumannii and Escherichia coli (E. coli) isolates against this antibiotic. In conclusion, our findings demonstrate that OMVs from highly drug-resistant CRAb confer protection against last-resort antibiotics to non-resistant bacterial pathogens.
Collapse
Affiliation(s)
- Rodrigo Monteiro
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands; Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Beatriz Santamarina Alcantud
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands
| | - Sjouke Piersma
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands
| | - Antoni P A Hendrickx
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Sandra Maaß
- University of Greifswald, Centre of Functional Genomics of Microbes, Department of Microbial Proteomics, Institute of Microbiology, Greifswald, Germany
| | - Dörte Becher
- University of Greifswald, Centre of Functional Genomics of Microbes, Department of Microbial Proteomics, Institute of Microbiology, Greifswald, Germany
| | - Joana Azeredo
- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Erik Bathoorn
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands
| | - Jan Maarten van Dijl
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands.
| |
Collapse
|
4
|
Qin Z, Li Y, Shao X, Li K, Bai Y, Wang B, Ma F, Shi W, Song L, Zhuang A, He F, Ding C, Yang W. HNF4A functions as a hepatocellular carcinoma oncogene or tumor suppressor depending upon the AMPK pathway activity status. Cancer Lett 2025; 623:217732. [PMID: 40254090 DOI: 10.1016/j.canlet.2025.217732] [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: 05/28/2024] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Cancer cells frequently undergo energy metabolic stress induced by the increased dynamics of nutrient supply. Hepatocyte nuclear factor 4A (HNF4A) is a master transcription factor (TF) in hepatocytes that regulates metabolism and differentiation. However, the mechanism underlying how HNF4A functions in cancer progression remains unclear due to conflicting results observed in numerous studies. To address the roles of HNF4A in hepatocellular carcinoma (HCC), we investigated the regulatory functions of HNF4A in HCC cells under different glucose supply conditions. We found that HNF4A exhibited tumor-suppressive effects on the proliferation and migration of HCC cells in glucose-sufficient conditions and tumor-promotive effects on HCC cells in glucose-insufficient conditions. Further investigation revealed that this diverse function of HNF4A was dependent upon the AMPK pathway activity. Similarly, the prognosis predicted by HNF4A was also correlated with whether the AMPKa expression levels were low or high in clinical HCC patients. Multiomics approaches consisting of proteomics and ChIP-seq revealed that key HNF4A target genes, including NEDD4 and RPS6KA2, are involved in the diverse function of HNF4A in HCC in response to the AMPK activity status. Specifically, HNF4A could bind to the promoter region of NEDD4 and RPS6KA2, and upregulating their expression. Our study has demonstrated the relationship between and synergism of AMPK and HNF4A in the progression of HCC under diverse nutrient conditions.
Collapse
Affiliation(s)
- Zhaoyu Qin
- Department of Pediatric Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China; State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Yan Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Xiexiang Shao
- Department of Pediatric Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Kai Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Yihe Bai
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Bing Wang
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Fahan Ma
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Wenhao Shi
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China; School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Lei Song
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Aojia Zhuang
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China
| | - Fuchu He
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China; School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Chen Ding
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Institutes of Biomedical Sciences, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai 200032, China; State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, National Center for Protein Sciences (The PHOENIX Center, Beijing), Beijing, 102206, China
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
5
|
Aguilera LU, Weber LM, Ron E, King CR, Öcal K, Popinga A, Cook J, May MP, Raymond WS, Fox ZR, Forero-Quintero LS, Forman JR, David A, Munsky B. Methods in quantitative biology-from analysis of single-cell microscopy images to inference of predictive models for stochastic gene expression. Phys Biol 2025; 22:042001. [PMID: 40388970 DOI: 10.1088/1478-3975/adda85] [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: 03/23/2025] [Accepted: 05/19/2025] [Indexed: 05/21/2025]
Abstract
The field of quantitative biology (q-bio) seeks to provide precise and testable explanations for observed biological phenomena by applying mathematical and computational methods. The central goals of q-bio are to (1) systematically propose quantitative hypotheses in the form of mathematical models, (2) demonstrate that these models faithfully capture a specific essence of a biological process, and (3) correctly forecast the dynamics of the process in new, and previously untested circumstances. Achieving these goals depends on accurate analysis and incorporating informative experimental data to constrain the set of potential mathematical representations. In this introductory tutorial, we provide an overview of the state of the field and introduce some of the computational methods most commonly used in q-bio. In particular, we examine experimental techniques in single-cell imaging, computational tools to process images and extract quantitative data, various mechanistic modeling approaches used to reproduce these quantitative data, and techniques for data-driven model inference and model-driven experiment design. All topics are presented in the context of additional online resources, including open-source Python notebooks and open-ended practice problems that comprise the technical content of the annual Undergraduate Quantitative Biology Summer School (UQ-Bio).
Collapse
Affiliation(s)
- Luis U Aguilera
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
- Department of Biochemistry and Molecular Genetics, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Lisa M Weber
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
- School of Mathematics and Engineering, Front Range Community College, Fort Collins, CO 80526, United States of America
| | - Eric Ron
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
| | - Connor R King
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
- Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO 80523, United States of America
| | - Kaan Öcal
- School of BioSciences, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Alex Popinga
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
- School of Biological Sciences, University of Auckland, Auckland CBD, Auckland 1010, New Zealand
| | - Joshua Cook
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
| | - Michael P May
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
| | - William S Raymond
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
| | - Zachary R Fox
- Oak Ridge National Laboratory, Oak Ridge, TN 37830, United States of America
| | - Linda S Forero-Quintero
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
| | - Jack R Forman
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
| | - Alexandre David
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
| | - Brian Munsky
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, United States of America
| |
Collapse
|
6
|
Gonzalez-Magaldi M, Gullapalli A, Papoulas O, Liu C, Leung AYH, Guo L, Brilot AF, Marcotte EM, Ke Z, Leahy DJ. Structure and organization of full-length epidermal growth factor receptor in extracellular vesicles by cryo-electron tomography. Proc Natl Acad Sci U S A 2025; 122:e2424678122. [PMID: 40455995 DOI: 10.1073/pnas.2424678122] [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: 11/25/2024] [Accepted: 04/22/2025] [Indexed: 06/11/2025] Open
Abstract
We report here transport of full-length epidermal growth factor receptor (EGFR), Insulin Receptor, 7-pass transmembrane receptor Smoothened, and 13-pass Sodium-iodide symporter to extracellular vesicles (EVs) for structural and functional studies. Mass spectrometry confirmed the transported proteins are the most abundant in EV membranes, and the presence of many receptor-interacting proteins in EVs demonstrates their utility for characterizing membrane protein interactomes. Cryo-electron tomography of EGFR-containing EVs reveals that EGFR forms clusters in both the presence and absence of EGF with a ~3 nm gap between the inner membrane and cytoplasmic density. EGFR extracellular region (ECR) dimers do not form regular arrays in these clusters. Subtomogram averaging of the 150 kDa EGF-bound EGFR ECR dimer yielded a 15 Å map into which the crystal structure of the ligand-bound EGFR ECR dimer fits well. These findings refine our understanding of EGFR activation, clustering, and signaling and establish EVs as a versatile platform for structural and functional characterization of human membrane proteins in cell-derived membranes.
Collapse
Affiliation(s)
| | - Anuradha Gullapalli
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Ophelia Papoulas
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Chang Liu
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Adelaide Y-H Leung
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Luqiang Guo
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Axel F Brilot
- Center for Biomedical Research Support, The University of Texas at Austin, Austin, TX 78712
| | - Edward M Marcotte
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| | - Zunlong Ke
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
- LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, TX 78712
| | - Daniel J Leahy
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712
| |
Collapse
|
7
|
Zhuang S, Diao X, Xiang R, Huang J, Pang J, Lu N, Liang Q, Pan X, Liu R, Fang C, Liang X, Peng W, Zeng H. Dual-omics reveals temporal translational recovery landscapes and cryodamage repair mechanisms in vitrified mouse oocytes. J Assist Reprod Genet 2025:10.1007/s10815-025-03482-w. [PMID: 40493158 DOI: 10.1007/s10815-025-03482-w] [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: 01/10/2025] [Accepted: 04/10/2025] [Indexed: 06/12/2025] Open
Abstract
PURPOSE The efficacy of oocyte cryopreservation still requires enhancement. Current understanding of cryodamage is largely limited to the organelle level, and the impact of freezing on oocyte gene expression remains unclear. METHODS In this study, we employed an innovative dual-omics approach to assess the transcriptional and translational profiles of mouse metaphase II (MII) oocytes during the initial 4 h post-thaw. RESULTS Our mapping of the translational recovery in vitrified mouse oocytes post-thaw revealed a critical 2-h window that is optimal for recovery. We confirmed the mitochondrial damage associated with vitrification and identified the activation of autophagy and proteasomal degradation during this period. Additionally, our analysis indicates that vitrified oocytes have another repair response to counteract cryoinjury, involving spindle remodeling and membrane recycling. CONCLUSIONS These findings can guide future efforts to improve oocyte vitrification outcomes by improving repair processes and not focusing solely on the damage processes.
Collapse
Affiliation(s)
- Shaohong Zhuang
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoting Diao
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Rui Xiang
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jiana Huang
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jiahui Pang
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Na Lu
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiqi Liang
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xinyi Pan
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Rang Liu
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Cong Fang
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyan Liang
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wenlin Peng
- Incita Fertility Center, Phnom Penh, Cambodia.
| | - Haitao Zeng
- Reproductive Medicine Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
- Guangdong Engineering Technology Research Center of Fertility Preservation, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
8
|
Douvris A, Maadelat A, Porter CJ, Burger D, Burns KD. miR-486-5p Inhibits eNOS and Angiogenesis in Cultured Endothelial Cells by Targeting MAML3. J Cell Mol Med 2025; 29:e70589. [PMID: 40432288 PMCID: PMC12116925 DOI: 10.1111/jcmm.70589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/22/2025] [Accepted: 04/26/2025] [Indexed: 05/29/2025] Open
Abstract
Kidney ischemia-reperfusion (I/R) is associated with endothelial injury. Administration of miRNA (miR)-486-5p protects against rat kidney I/R injury, with localisation to capillary endothelial cells, although it inhibits I/R-induced endothelial nitric oxide synthase (eNOS) protein expression. Here, we studied the effect of miR-486-5p on eNOS and endothelial cell function and determined its mRNA targets. Human umbilical vein endothelial cells (HUVECs) were transfected with the miR-486-5p mimic and assayed for proliferation, migration and network formation. Biotinylated miR-486-5p was transfected for pulldown of bound mRNA, followed by RNA sequencing. miR-486-5p markedly decreased eNOS mRNA and protein in HUVECs (p < 0.001) and decreased eNOS protein in human pulmonary microvascular endothelial cells (p < 0.05), although eNOS was not a direct target of miR-486-5p. miR-486-5p inhibited angiogenesis, which was rescued with eNOS plasmid transfection. RNA sequencing of biotinylated miR-486-5p pulldown RNA revealed highly significant enrichment in predicted targets FOXO1, FOXP1, TNFSF4, MAML3 and CELSR3, and in the non-predicted target SPCS2. RT-qPCR validated these transcripts as inhibited by miR-486-5p. While silencing of FOXO1 had no impact on eNOS protein, MAML3 silencing inhibited eNOS levels. miR-486-5p inhibits angiogenesis in endothelial cells via eNOS down-regulation, which involves selective targeting of MAML3. These data support a novel pathway regulating endothelial cell function.
Collapse
Affiliation(s)
- Adrianna Douvris
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research InstituteUniversity of OttawaOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Ali Maadelat
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research InstituteUniversity of OttawaOttawaOntarioCanada
| | - Christopher J. Porter
- Ottawa Bioinformatics Core FacilityOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Dylan Burger
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research InstituteUniversity of OttawaOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Kevin D. Burns
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research InstituteUniversity of OttawaOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
9
|
Sharma N, Sharma N, Biswas A, Gupta S, Behura A, Rodriguez GM. Iron-restricted Mycobacterium tuberculosis exports pathogenicity factors packed in extracellular vesicles. PLoS One 2025; 20:e0324919. [PMID: 40445943 PMCID: PMC12124568 DOI: 10.1371/journal.pone.0324919] [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: 01/20/2025] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
Mycobacterium tuberculosis, the pathogen responsible for human tuberculosis, responds to iron limitation by increasing the production of extracellular vesicles. This study examined the protein composition of induced M. tuberculosis extracellular membrane vesicles using chromatography coupled with mass spectrometry. The results revealed that vesicles contain key pathogenicity factors, including proteins that enhance bacterial survival, immune evasion, and inflammation. These findings deepen our understanding of the potential role of extracellular vesicles in M. tuberculosis-host interactions. The data can also aid in identifying new biomarkers of infection and developing vesicle-based, culture-independent TB diagnostic platforms.
Collapse
Affiliation(s)
- Nishant Sharma
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Nevadita Sharma
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Ashis Biswas
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Shamba Gupta
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Assirbad Behura
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Gloria Marcela Rodriguez
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| |
Collapse
|
10
|
Wang G, Zhao J, Lin Y, Liu T, Zhao Y, Zhao H. scMODAL: a general deep learning framework for comprehensive single-cell multi-omics data alignment with feature links. Nat Commun 2025; 16:4994. [PMID: 40442129 PMCID: PMC12122792 DOI: 10.1038/s41467-025-60333-z] [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: 03/30/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
Recent advancements in single-cell technologies have enabled comprehensive characterization of cellular states through transcriptomic, epigenomic, and proteomic profiling at single-cell resolution. These technologies have significantly deepened our understanding of cell functions and disease mechanisms from various omics perspectives. As these technologies evolve rapidly and data resources expand, there is a growing need for computational methods that can integrate information from different modalities to facilitate joint analysis of single-cell multi-omics data. However, integrating single-cell omics datasets presents unique challenges due to varied feature correlations and technology-specific limitations. To address these challenges, we introduce scMODAL, a deep learning framework tailored for single-cell multi-omics data alignment using feature links. scMODAL integrates datasets with limited known positively correlated features, leveraging neural networks and generative adversarial networks to align cell embeddings and preserve feature topology. Our experiments demonstrate scMODAL's effectiveness in removing unwanted variation, preserving biological information, and accurately identifying cell subpopulations across diverse datasets. scMODAL not only advances integration tasks but also supports downstream analyses such as feature imputation and feature relationship inference, offering a robust solution for advancing single-cell multi-omics research.
Collapse
Affiliation(s)
- Gefei Wang
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Jia Zhao
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Yingxin Lin
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Tianyu Liu
- Department of Biostatistics, Yale University, New Haven, CT, USA
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Yize Zhao
- Department of Biostatistics, Yale University, New Haven, CT, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale University, New Haven, CT, USA.
- Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
| |
Collapse
|
11
|
Morton-Hayward A, Flannery S, Vendrell I, Fischer R. Deep palaeoproteomic profiling of archaeological human brains. PLoS One 2025; 20:e0324246. [PMID: 40435004 PMCID: PMC12118856 DOI: 10.1371/journal.pone.0324246] [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: 01/10/2025] [Accepted: 04/22/2025] [Indexed: 06/01/2025] Open
Abstract
Palaeoproteomics leverages the persistence, diversity, and biological import of ancient proteins to explore the past, and answer fundamental questions about phylogeny, environment, diet, and disease. These insights are largely gleaned from hard tissues like bone and teeth, as well-established protocols exist for extracting ancient proteins from mineralised tissues. No such method, however, exists for the soft tissues, which are underexplored in palaeoproteomics given permission for destructive analysis routinely depends on a proven methodology. Considering less than one-tenth of all human proteins are expressed in bone, compared to three-quarters in the internal organs, the amount of biological information presently inaccessible is substantial. We address this omission with an optimised LC-FAIMS-MS/MS workflow yielding the largest, most diverse palaeoproteome yet described. Using archaeological human brains, we test ten protocols with varied chemistries and find that urea lysis effectively disrupts preserved membrane regions to expose low-abundant, intracellular analytes. Further, we show that ion mobility spectrometry improves unique protein identification by as much as 40%, and represents a means of "cleaning" dirty archaeological samples. Our methodology will be useful for improving protein recovery from a range of ancient tissues and depositional environments.
Collapse
Affiliation(s)
- Alexandra Morton-Hayward
- Department of Earth Sciences, University of Oxford, Oxford, United Kingdom
- Target Discovery Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Sarah Flannery
- Department of Earth Sciences, University of Oxford, Oxford, United Kingdom
| | - Iolanda Vendrell
- Department of Earth Sciences, University of Oxford, Oxford, United Kingdom
| | - Roman Fischer
- Department of Earth Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
12
|
Gao Y, Guo L, Shi G, Wang R, Wang X, Lou J. Emerging roles of ribosome translation in stem cells and stem cell therapy - a review. Cell Biosci 2025; 15:71. [PMID: 40437562 PMCID: PMC12121016 DOI: 10.1186/s13578-025-01412-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/16/2025] [Indexed: 06/01/2025] Open
Abstract
Stem cells differ from other somatic cells in that they possess self-renewal and differentiation potential, which endows them with unique characteristics, and have great therapeutic potential. Studies have shown that the self-renewal and differentiation potential of stem cells is regulated by ribosomes during protein synthesis. In this review, we discuss the translation regulation mechanisms and ribosome biogenesis in stem cells. Protein translation levels and ribosome biogenesis change dynamically during the development and differentiation of stem cells, and hierarchical translational regulation promotes stem cell differentiation. We also demonstrate that mitochondrial protein translation plays an important role in the regulation of stem cell fate. Ribosomes not only mediate the self-renewal and differentiation of stem cells through protein synthesis. They are also a key target for stem cell therapy. Understanding the mechanism of ribosome regulation in stem cells will allow better control of stem cells for their application.
Collapse
Affiliation(s)
- Yanyan Gao
- Department of Anesthesiology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, No. 99 Long Cheng Road, Taiyuan, 030032, China.
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Linlin Guo
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Gaoxiang Shi
- Department of Anesthesiology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, No. 99 Long Cheng Road, Taiyuan, 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ruifang Wang
- Department of Anesthesiology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, No. 99 Long Cheng Road, Taiyuan, 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xu'an Wang
- Department of Anesthesiology, Tongji Shanxi Hospital, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, No. 99 Long Cheng Road, Taiyuan, 030032, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jizhong Lou
- Laboratory of RNA Biology, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
13
|
Marintchev A. Exploring the interaction dynamics of eukaryotic translation initiation factor 2. Biochem Soc Trans 2025:BST20253022. [PMID: 40411218 DOI: 10.1042/bst20253022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 05/07/2025] [Indexed: 05/26/2025]
Abstract
Eukaryotic translation initiation typically involves recruitment of the 43S ribosomal pre-initiation complex (PIC) to the 5'-end of the mRNA to form the 48S PIC, followed by scanning in search of a start codon in a favorable nucleotide complex. The start codon is recognized through base-pairing with the anticodon of the initiator Met-tRNAi. The stringency of start codon selection controls the probability of initiation from a start codon in a suboptimal nucleotide context. Met-tRNAi itself is recruited to the 43S PIC by the eukaryotic translation initiation factor 2 (eIF2), in the form of the eIF2-GTP•Met-tRNAi ternary complex (TC). GTP hydrolysis by eIF2, promoted by its GTPase-activating protein eIF5, leads to the release of eIF2-GDP from the PIC. Recycling of eIF2-GDP to TC is promoted by the guanine nucleotide exchange factor eIF2B. Its inhibition by a number of stress factors triggers the integrated stress response (ISR). This review describes the recent advances in elucidating the interactions of eIF2 and its partners, with an emphasis on the timing and dynamics of their binding to, and release from the PIC. Special attention is given to the regulation of the stringency of start codon selection and the ISR. The discussion is mostly limited to translation initiation in mammals and budding yeast.
Collapse
Affiliation(s)
- Assen Marintchev
- Department of Pharmacology, Physiology, & Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, U.S.A
| |
Collapse
|
14
|
Zhang Z, Fan YN, Jiang SQ, Ma YJ, Yu YR, Qing YX, Li QR, Liu YL, Shen S, Wang J. Recent Advances in mRNA Delivery Systems for Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e17571. [PMID: 40391789 DOI: 10.1002/advs.202417571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 04/01/2025] [Indexed: 05/22/2025]
Abstract
mRNA therapy is a promising approach in oncology, offering innovative applications such as tumor vaccines, protein replacement therapy, cell therapy, and gene therapy. However, challenges such as mRNA stability and delivery efficiency must be addressed. Advances in delivery system technologies are crucial for precise mRNA delivery, enhancing treatment safety and efficacy. The development of delivery systems requires accurate organ or cell targeting, intelligent release mechanisms, and optimized administration routes. This review outlines the applications of mRNA therapy in oncology, as well as the utilization of nonviral vectors, encompassing organic, inorganic, and biomimetic systems. It further elucidates the strategies for passive and active vector targeting and examines recent advances in the realm of stimuli-responsive delivery systems that are sensitive to pH and ultrasound. Additionally, the review addresses the development of noninvasive mRNA delivery systems designed for oral and pulmonary administration. The current challenges and emerging trends of mRNA therapy are discussed, and the potential strategies to mitigate these issues are emphasized.
Collapse
Affiliation(s)
- Zheng Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Ya-Nan Fan
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Si-Qi Jiang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Ya-Jing Ma
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Yao-Ru Yu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Yu-Xin Qing
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Qian-Ru Li
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Yi-Lin Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Song Shen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Jun Wang
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| |
Collapse
|
15
|
Farrell C, Buhidma Y, Mumford P, Heywood WE, Hällqvist J, Flores-Aguilar L, Andrews EJ, Rahimzadah N, Taso OS, Doran E, Swarup V, Head E, Lashley T, Mills K, Toomey CE, Wiseman FK. Apolipoprotein E abundance is elevated in the brains of individuals with Down syndrome-Alzheimer's disease. Acta Neuropathol 2025; 149:49. [PMID: 40387921 PMCID: PMC12089208 DOI: 10.1007/s00401-025-02889-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/25/2025] [Accepted: 05/01/2025] [Indexed: 05/20/2025]
Abstract
Trisomy of chromosome 21, the cause of Down syndrome (DS), is the most commonly occurring genetic cause of Alzheimer's disease (AD). Here, we compare the frontal cortex proteome of people with Down syndrome-Alzheimer's disease (DSAD) to demographically matched cases of early onset AD and healthy ageing controls. We find dysregulation of the proteome, beyond proteins encoded by chromosome 21, including an increase in the abundance of the key AD-associated protein, APOE, in people with DSAD compared to matched cases of AD. To understand the cell types that may contribute to changes in protein abundance, we undertook a matched single-nuclei RNA-sequencing study, which demonstrated that APOE expression was elevated in subtypes of astrocytes, endothelial cells, and pericytes in DSAD. We further investigate how trisomy 21 may cause increased APOE. Increased abundance of APOE may impact the development of, or response to, AD pathology in the brain of people with DSAD, altering disease mechanisms with clinical implications. Overall, these data highlight that trisomy 21 alters both the transcriptome and proteome of people with DS in the context of AD, and that these differences should be considered when selecting therapeutic strategies for this vulnerable group of individuals who have high risk of early onset dementia.
Collapse
Affiliation(s)
- Clíona Farrell
- UK Dementia Research Institute at University College London, London, UK
- Queen Square Institute of Neurology, University College London, London, UK
| | - Yazead Buhidma
- Queen Square Institute of Neurology, University College London, London, UK
| | - Paige Mumford
- UK Dementia Research Institute at University College London, London, UK
- Queen Square Institute of Neurology, University College London, London, UK
| | - Wendy E Heywood
- UCL Great Ormond Street Institute of Child Heath, University College London, London, UK
| | - Jenny Hällqvist
- UCL Great Ormond Street Institute of Child Heath, University College London, London, UK
| | - Lisi Flores-Aguilar
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Elizabeth J Andrews
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Negin Rahimzadah
- Mathematical, Computational, and Systems Biology (MCSB) Program, University of California, Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems (CCBS), University of California Irvine, Irvine, CA, USA
| | - Orjona Stella Taso
- UK Dementia Research Institute at University College London, London, UK
- Queen Square Institute of Neurology, University College London, London, UK
| | - Eric Doran
- Department of Pediatrics, School of Medicine, University of California, Irvine, Orange, CA, USA
| | - Vivek Swarup
- Institute for Memory Impairments and Neurological Disorders (MIND), University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems (CCBS), University of California Irvine, Irvine, CA, USA
- Department of Neurobiology and Behaviour, University of California, Irvine, CA, USA
| | - Elizabeth Head
- Department of Pathology and Laboratory Medicine, University of California, Irvine, CA, USA
| | - Tammaryn Lashley
- Queen Square Institute of Neurology, University College London, London, UK
| | - Kevin Mills
- UCL Great Ormond Street Institute of Child Heath, University College London, London, UK
| | - Christina E Toomey
- Queen Square Institute of Neurology, University College London, London, UK.
- The Francis Crick Institute, London, UK.
| | - Frances K Wiseman
- UK Dementia Research Institute at University College London, London, UK.
- Queen Square Institute of Neurology, University College London, London, UK.
| |
Collapse
|
16
|
Rubtsova M, Mokrushina Y, Andreev D, Poteshnova M, Shepelev N, Koryagina M, Moiseeva E, Malabuiok D, Prokopenko Y, Terekhov S, Chernov A, Vodovozova E, Smirnov I, Dontsova O, Gabibov A, Rubtsov Y. A Luciferase-Based Approach for Functional Screening of 5' and 3' Untranslated Regions of the mRNA Component for mRNA Vaccines. Vaccines (Basel) 2025; 13:530. [PMID: 40432139 PMCID: PMC12115628 DOI: 10.3390/vaccines13050530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 05/05/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: The recent COVID-19 pandemic caused by SARS-CoV-2 infection has highlighted the need for protocols for rapid development of efficient screening methods to search for the optimal mRNA vaccine structures against mutable viral agents. The unmatched success of mRNA vaccines by Pfizer and Moderna encoding the spike protein of SARS-CoV-2 confirms the potential of lipid nanoparticles for mRNA delivery for an accelerated development of new vaccines. The efficacy of vaccination and the production cost of mRNA-based vaccines largely depend on the composition of mRNA components, since the synthesis of an immunogenic protein requires precise and efficient translation in vivo. The composition of 5' and 3' UTR combinations of mRNA has a strong impact on the translation efficiency. The major objective of this study was to increase the probability of producing the immunogenic protein encoded by vaccine mRNA. For this purpose, we proposed to find a new combination of natural UTRs and, in parallel with that, to design and test the system for in vivo selection of translationally active UTRs. Methods: By using Ribo-Seq analysis, sets of candidate short UTRs were generated. These UTRs were tested both in cell cultures and in mice for effective production of secreted nanoluciferase (NLuc) and the S protein of SARS-CoV-2. A combination of the most effective UTRs was used to generate a prototype of an mRNA vaccine capable of inducing neutralizing antibodies against coronavirus. Results: The usefulness of the selected UTRs for vaccine development was tested by implicating the full-length coding sequence of SARS-CoV-2 S protein to produce the main immunogen. As a result, the system for functional screening of UTRs was created by using the NLuc gene. Conclusions: The proposed approach allows non-invasive quantitative assessment of the translational activity of UTRs in the blood serum of mice. By using the full-length sequence of SARS-CoV-2 S protein as a prototype, we demonstrated that the combination of UTRs selected using our luciferase-based reporter assay induces IgG titers and neutralization rates comparable to those obtained by using UTRs from commercial S-protein-based mRNA vaccines.
Collapse
Affiliation(s)
- Maria Rubtsova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (M.P.); (N.S.); (M.K.)
| | - Yuliana Mokrushina
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| | - Dmitry Andreev
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| | - Maria Poteshnova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (M.P.); (N.S.); (M.K.)
| | - Nikita Shepelev
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (M.P.); (N.S.); (M.K.)
| | - Mariya Koryagina
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (M.P.); (N.S.); (M.K.)
| | - Ekaterina Moiseeva
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| | - Diana Malabuiok
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| | - Yury Prokopenko
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| | - Stanislav Terekhov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| | - Aleksander Chernov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| | - Elena Vodovozova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| | - Ivan Smirnov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (M.P.); (N.S.); (M.K.)
- Endocrinology Research Center of the Ministry of Health of the Russian Federation, 117292 Moscow, Russia
| | - Olga Dontsova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia; (M.P.); (N.S.); (M.K.)
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 143025 Moscow, Russia
| | - Alexander Gabibov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| | - Yury Rubtsov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Science, 117997 Moscow, Russia; (M.R.); (Y.M.); (D.A.); (E.M.); (D.M.); (Y.P.); (S.T.); (A.C.); (E.V.); (I.S.); (O.D.)
| |
Collapse
|
17
|
Nagel M, Taatjes DJ. Regulation of RNA polymerase II transcription through re-initiation and bursting. Mol Cell 2025; 85:1907-1919. [PMID: 40378829 DOI: 10.1016/j.molcel.2025.04.011] [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: 12/20/2024] [Revised: 03/13/2025] [Accepted: 04/07/2025] [Indexed: 05/19/2025]
Abstract
The regulation of RNA polymerase II (RNAPII) activity requires orchestrated responses among genomic regulatory sequences and an expansive set of proteins and protein complexes. Despite intense study over five decades, mechanistic insights continue to emerge. Within the past 10 years, live-cell imaging and single-cell transcriptomics experiments have yielded new information about enhancer-promoter communication, transcription factor dynamics, and the kinetics of RNAPII transcription activation. These insights have established RNAPII re-initiation and bursting as a common regulatory phenomenon with widespread implications for gene regulation in health and disease. Here, we summarize regulatory strategies that help control RNAPII bursting in eukaryotic cells, which is defined as short periods of active transcription followed by longer periods of inactivity. We focus on RNAPII re-initiation (i.e., a "burst" of two or more polymerases that initiate from the same promoter), with an emphasis on molecular mechanisms, open questions, and controversies surrounding this distinct regulatory stage.
Collapse
Affiliation(s)
- Michael Nagel
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA
| | - Dylan J Taatjes
- Department of Biochemistry, University of Colorado, Boulder, CO 80303, USA.
| |
Collapse
|
18
|
Trendel J, Trendel S, Sha S, Greulich F, Goll S, Wudy SI, Kleigrewe K, Kubicek S, Uhlenhaut NH, Kuster B. The human proteome with direct physical access to DNA. Cell 2025:S0092-8674(25)00507-0. [PMID: 40409270 DOI: 10.1016/j.cell.2025.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 01/10/2025] [Accepted: 04/27/2025] [Indexed: 05/25/2025]
Abstract
In a human cell, DNA is packed with histones, RNA, and chromatin-associated proteins, forming a cohesive gel. At any given moment, only a subset of the proteome has physical access to the DNA and organizes its structure, transcription, replication, repair, and other essential molecular functions. We have developed a "zero-distance" photo-crosslinking approach to quantify proteins in direct contact with DNA in living cells. Collecting DNA interactomes from human breast cancer cells, we present an atlas of over one thousand proteins with physical access to DNA and hundreds of peptide-nucleotide crosslinks pinpointing protein-DNA interfaces with single-amino-acid resolution. Quantitative comparisons of DNA interactomes from differentially treated cells recapitulate the recruitment of key transcription factors as well as DNA repair proteins and uncover fast-acting restrictors of chromatin accessibility on a timescale of minutes. This opens a direct way to explore genomic regulation in a hypothesis-free manner, applicable to many organisms and systems.
Collapse
Affiliation(s)
- Jakob Trendel
- Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich (TUM), Freising, Germany
| | | | - Shuyao Sha
- Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich (TUM), Freising, Germany
| | - Franziska Greulich
- Metabolic Programming, TUM School of Life Sciences, ZIEL-Institute for Food & Health, Technical University of Munich (TUM), Freising, Germany
| | - Sandra Goll
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Susanne I Wudy
- Bavarian Center for Biomolecular Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich (TUM), Freising, Germany
| | - Karin Kleigrewe
- Bavarian Center for Biomolecular Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich (TUM), Freising, Germany
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - N Henriette Uhlenhaut
- Metabolic Programming, TUM School of Life Sciences, ZIEL-Institute for Food & Health, Technical University of Munich (TUM), Freising, Germany; Institute for Diabetes and Obesity (IDO) & Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich (HMGU) and German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Bernhard Kuster
- Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich (TUM), Freising, Germany.
| |
Collapse
|
19
|
Li W, Xie R, Chen H, Lin J, Zhong M, Zhang J, Zheng S, Jiang C, Chen X, Xu S. METTL1-mediated m 7G tRNA modification drives papillary thyroid cancer progression and metastasis by regulating the codon-specific translation of TNF-α. Cell Death Dis 2025; 16:378. [PMID: 40360483 PMCID: PMC12075834 DOI: 10.1038/s41419-025-07716-8] [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: 12/10/2024] [Revised: 04/16/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025]
Abstract
N7-methylguanosine (m7G) modification of transfer RNA (tRNA) is essential for the biological functions of tRNAs and has been found to play a regulatory role in a variety of human cancers. However, the biological function of METTL1-mediated m7G tRNA modification in papillary thyroid cancer (PTC) is unclear. Here, we found that METTL1 is significantly upregulated in PTC tissues compared to normal control tissues and is associated with poor PTC prognosis. Functional analysis confirmed that METTL1 promotes the proliferation and metastasis of PTC cells in a manner dependent on its tRNA methyltransferase activity. Mechanistically, METTL1 knockdown leads to a decrease in the abundance of certain m7G-modified tRNAs, which suppresses the m7G tRNA modification-mediated codon-specific translation of TNF-α. Furthermore, exogenous supplementation with TNF-α partially reversed the decrease in the proliferation and metastasis of PTC cells induced by METTL1 deletion. Positive correlations between METTL1, WDR4, and TNF-α expression, which affect the proliferation and metastasis of PTC, were confirmed via analysis of microarrays containing PTC tissues. These results demonstrate the oncogenic role of METTL1-mediated m7G tRNA modification in regulating codon-specific translation efficiency in PTC and suggest that targeting METTL1 may be a promising therapeutic approach for overcoming PTC progression by inhibiting PTC cell proliferation and metastasis.
Collapse
Affiliation(s)
- Weiwei Li
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ruiwang Xie
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Huaying Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Junyu Lin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Minjie Zhong
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Junsi Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shengkai Zheng
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Cen Jiang
- Central Laboratory, Fujian Medical University Union Hospital, Fuzhou, China
| | - Xiangjin Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Sunwang Xu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Thyroid and Breast Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, Fuzhou, China.
| |
Collapse
|
20
|
Abdul-Khalek N, Picciani M, Shouman O, Wimmer R, Overgaard MT, Wilhelm M, Gregersen Echers S. To Fly, or Not to Fly, That Is the Question: A Deep Learning Model for Peptide Detectability Prediction in Mass Spectrometry. J Proteome Res 2025. [PMID: 40344201 DOI: 10.1021/acs.jproteome.4c00973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Identifying detectable peptides, known as flyers, is key in mass spectrometry-based proteomics. Peptide detectability is strongly related to peptide sequences and their resulting physicochemical properties. Moreover, the high variability in MS data challenges the development of a generic model for detectability prediction, underlining the need for customizable tools. We present Pfly, a deep learning model developed to predict peptide detectability based solely on peptide sequence. Pfly is a versatile and reliable state-of-the-art tool, offering high performance, accessibility, and easy customizability for end-users. This adaptability allows researchers to tailor Pfly to specific experimental conditions, improving accuracy and expanding applicability across various research fields. Pfly is an encoder-decoder with an attention mechanism, classifying peptides as flyers or non-flyers, and providing both binary and categorical probabilities for four distinct classes defined in this study. The model was initially trained on a synthetic peptide library and subsequently fine-tuned with a biological dataset to mitigate bias toward synthesizability, improving predictive capacity and outperforming state-of-the-art predictors in benchmark comparisons across different human and cross-species datasets. The study further investigates the influence of protein abundance and rescoring, illustrating the negative impact on peptide identification due to misclassification. Pfly has been integrated into the DLOmix framework and is accessible on GitHub at https://github.com/wilhelm-lab/dlomix.
Collapse
Affiliation(s)
- Naim Abdul-Khalek
- Department of Chemistry and Bioscience, Aalborg University, 9220 Aalborg, Denmark
| | - Mario Picciani
- Computational Mass Spectrometry, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Omar Shouman
- Computational Mass Spectrometry, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Reinhard Wimmer
- Department of Chemistry and Bioscience, Aalborg University, 9220 Aalborg, Denmark
| | | | - Mathias Wilhelm
- Computational Mass Spectrometry, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
- Munich Data Science Institute, Technical University of Munich, 85748 Garching, Germany
| | | |
Collapse
|
21
|
Tsoy S, Liu J. Regulation of Protein Synthesis at the Translational Level: Novel Findings in Cardiovascular Biology. Biomolecules 2025; 15:692. [PMID: 40427584 PMCID: PMC12108789 DOI: 10.3390/biom15050692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Translational regulation plays a pivotal role in cardiac gene expression, influencing protein synthesis in response to physiological and pathological stimuli. Although transcription regulates gene expression, translation ultimately determines protein levels, making it a crucial research focus. In cardiomyocytes, disruptions in this process contribute to various cardiac diseases, including hypertrophy, fibrosis, dilated cardiomyopathy, ischemic heart disease, and diabetic cardiomyopathy. Emerging evidence highlights the significance of translational regulation across multiple cardiac cell types, such as cardiomyocytes and fibroblasts, and its role in disease progression. During cardiac remodeling, transcriptomic changes are often modest, suggesting that post-transcriptional mechanisms, particularly translation, play a dominant role in cellular adaptation. This review discusses key methodologies for studying translational regulation and novel mechanisms of translational regulation related to different cardiac pathologies and highlights relevant therapeutic avenues for targeting these pathways.
Collapse
Affiliation(s)
- Sergey Tsoy
- Medical Scientist Training Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
22
|
Knoch D, Rugen N, Thiel J, Heuermann MC, Kuhlmann M, Rizzo P, Meyer RC, Wagner S, Schippers JHM, Braun HP, Altmann T. A spatio-temporal transcriptomic and proteomic dataset of developing Brassica napus seeds. Sci Data 2025; 12:759. [PMID: 40335559 PMCID: PMC12059035 DOI: 10.1038/s41597-025-05115-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Oilseed rape (Brassica napus) seeds are of major economic and nutritional value since they are rich in both oil and proteins, which accumulate predominantly in the embryonic cotyledons during the filling period. Developmental phases such as embryogenesis, seed filling, and maturation have been associated with specific changes in the transcriptional landscape and are controlled by interactions of regulatory components, particularly transcription factors and cis-regulatory elements. However, the global changes on the protein level remain largely elusive. Here, we investigated the dynamics of seed development by an integrative analysis of the seed transcriptome and proteome. Plants of the winter-type cultivar Express 617 were grown under controlled, field-like conditions in the IPK PhenoSphere, and developing seeds were collected for temporally and spatially resolved multi-omics analyses. The dataset covers five stages, from pre-storage to seed maturation, and includes spatial information on four dissected organs/tissues. It provides comprehensive insights into differentiation and developmental processes of the Brassica napus seed and may serve as starting point to select potentially important genes for detailed functional investigations.
Collapse
Affiliation(s)
- Dominic Knoch
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Department of Molecular Genetics, Corrensstraße 3, 06466 Seeland OT, Gatersleben, Germany
| | - Nils Rugen
- Institute of Plant Genetics, Leibniz University Hannover, Herrenhäuser Straße 2, 30419, Hannover, Germany
| | - Johannes Thiel
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Department of Molecular Genetics, Corrensstraße 3, 06466 Seeland OT, Gatersleben, Germany
| | - Marc C Heuermann
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Department of Molecular Genetics, Corrensstraße 3, 06466 Seeland OT, Gatersleben, Germany
| | - Markus Kuhlmann
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Department of Molecular Genetics, Corrensstraße 3, 06466 Seeland OT, Gatersleben, Germany
| | - Paride Rizzo
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Department of Molecular Genetics, Corrensstraße 3, 06466 Seeland OT, Gatersleben, Germany
| | - Rhonda C Meyer
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Department of Molecular Genetics, Corrensstraße 3, 06466 Seeland OT, Gatersleben, Germany
| | - Steffen Wagner
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Department of Molecular Genetics, Corrensstraße 3, 06466 Seeland OT, Gatersleben, Germany
| | - Jos H M Schippers
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Department of Molecular Genetics, Corrensstraße 3, 06466 Seeland OT, Gatersleben, Germany
| | - Hans-Peter Braun
- Institute of Plant Genetics, Leibniz University Hannover, Herrenhäuser Straße 2, 30419, Hannover, Germany
| | - Thomas Altmann
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK), Department of Molecular Genetics, Corrensstraße 3, 06466 Seeland OT, Gatersleben, Germany.
| |
Collapse
|
23
|
Luo Y, Fu Y, Kuang M, Wang J, Zhao R, Luo S, Wang L, Chen J, Xu S, Zhou C. Ciclosporin A potentiates venetoclax efficacy in FLT3-ITD AML by targeting the NFATC1-AKT-mTOR-BCL-2/MCL-1 signaling axis. Br J Haematol 2025. [PMID: 40328636 DOI: 10.1111/bjh.20137] [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: 01/05/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025]
Abstract
The Fms-like tyrosine kinase 3 internal tandem duplication (FLT3-ITD) mutation in acute myeloid leukaemia (AML) is associated with adverse clinical outcomes, including poor prognosis, high relapse rates and reduced responses to conventional treatment regimens. While venetoclax (VEN) monotherapy has shown limited efficacy in FLT3-ITD AML due to intrinsic resistance mechanisms, this study demonstrates that ciclosporin A (CsA) synergistically enhances VEN's anti-leukaemic activity. CsA significantly suppresses cell proliferation, induces mitochondrial apoptosis and impairs mitochondrial bioenergetics in FLT3-ITD AML cells. Mechanistically, CsA enhances the effects of VEN through the downregulation of NFATC1, a critical regulator of the PI3K/AKT/mTOR signalling pathway. This suppression of NFATC1 leads to the coordinated downregulation of the anti-apoptotic proteins BCL-2 and MCL-1, thereby overcoming resistance and reinstating therapeutic susceptibility in FLT3-ITD AML.
Collapse
Affiliation(s)
- Yu Luo
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yinghao Fu
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Mei Kuang
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jianming Wang
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Runlong Zhao
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Siqi Luo
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Wang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jieping Chen
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shuangnian Xu
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chengfang Zhou
- Department of Radiological Medicine, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Department of Hematology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
24
|
Wilczak M, Surman M, Jankowska U, Skupien-Rabian B, Przybyło M. MGAT3 and MGAT5 overexpression alters the protein cargo of extracellular vesicles released by metastatic melanoma cells. Biochem Biophys Res Commun 2025; 762:151749. [PMID: 40199132 DOI: 10.1016/j.bbrc.2025.151749] [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: 12/12/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
Extracellular vesicles (EVs) are potential non-invasive diagnostic, prognostic and therapeutic tools. Additionally, they are important contributors to tumorigenesis. Glycosylation has been found to modulate the composition of the EV proteome. Increased amounts of β1,6-branched N-glycans, synthesized by N-acetylglucosaminyltransferase V (GnT-V), are most commonly observed in melanoma and are associated with decreased cell adhesion and increased metastasis. The opposite effect is caused by the addition of bisecting GlcNAc by N-acetylglucosaminyltransferase III (GnT-III). To date, the impact of these enzymes on EV cargo in melanoma remains unexplored. Flow cytometry was used to study the surface glycosylation of genetic variants of WM266-4 melanoma cells with induced overexpression of GnT-III or GnT-V encoding genes (MGAT3 or MGAT5) and EVs released by these cells. LC-MS/MS proteomics was applied to analyze the effect of altered glycosylation on the proteome of released EVs, followed by detailed bioinformatic analysis. Flow cytometry analysis revealed dynamic changes in the surface glycosylation of EVs derived from melanoma cells overexpressing MGAT3 or MGAT5. Induced overexpression of MGAT3 or MGAT5 also caused significant changes in the proteome of EVs. The proteomic analysis identified a total of 1770 microvesicular and 704 exosomal proteins that play different roles in melanoma progression, including those with established diagnostic/prognostic potential and those closely associated with melanoma onset. Proteomic profiling of EVs derived from cells overexpressing MGAT3 and MGAT5 revealed functional changes in EV protein content driven by glycosylation modifications. The study presented a potential multifaced application of melanoma-derived EVs for diagnostic and prognostic purposes.
Collapse
Affiliation(s)
- Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387, Krakow, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University, 30-348, Krakow, Poland.
| | - Magdalena Surman
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387, Krakow, Poland.
| | - Urszula Jankowska
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387, Krakow, Poland.
| | - Bozena Skupien-Rabian
- Proteomics and Mass Spectrometry Core Facility, Malopolska Centre of Biotechnology, Jagiellonian University, 30-387, Krakow, Poland.
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, 30-387, Krakow, Poland.
| |
Collapse
|
25
|
Sabatier P, Lechner M, Guzmán UH, Beusch CM, Zeng X, Wang L, Izaguirre F, Seth A, Gritsenko O, Rodin S, Grinnemo KH, Ye Z, Olsen JV. Global analysis of protein turnover dynamics in single cells. Cell 2025; 188:2433-2450.e21. [PMID: 40168994 DOI: 10.1016/j.cell.2025.03.002] [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: 07/04/2024] [Revised: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 04/03/2025]
Abstract
Single-cell proteomics (SCPs) has advanced significantly, yet it remains largely unidimensional, focusing primarily on protein abundances. In this study, we employed a pulsed stable isotope labeling by amino acids in cell culture (pSILAC) approach to simultaneously analyze protein abundance and turnover in single cells (SC-pSILAC). Using a state-of-the-art SCP workflow, we demonstrated that two SILAC labels are detectable from ∼4,000 proteins in single HeLa cells recapitulating known biology. We performed a large-scale time-series SC-pSILAC analysis of undirected differentiation of human induced pluripotent stem cells (iPSCs) encompassing 6 sampling times over 2 months and analyzed >1,000 cells. Protein turnover dynamics highlighted differentiation-specific co-regulation of protein complexes with core histone turnover, discriminating dividing and non-dividing cells. Lastly, correlating cell diameter with the abundance of individual proteins showed that histones and some cell-cycle proteins do not scale with cell size. The SC-pSILAC method provides a multidimensional view of protein dynamics in single-cell biology.
Collapse
Affiliation(s)
- Pierre Sabatier
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden.
| | - Maico Lechner
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ulises H Guzmán
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christian M Beusch
- Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden; Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA 30322, USA
| | - Xinlei Zeng
- 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, China
| | - Longteng Wang
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | | | - Anjali Seth
- Cellenion SASU, 60F Avenue Rockefeller, 69008 Lyon, France
| | - Olga Gritsenko
- Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden
| | - Sergey Rodin
- Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden
| | - Karl-Henrik Grinnemo
- Cardio-Thoracic Translational Medicine (CTTM) Lab, Department of Surgical Sciences, Uppsala University, 752 37 Uppsala, Sweden
| | - Zilu Ye
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; 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, China.
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
26
|
Krouch D, Vreeke GJC, America AHP, Mes JJ, Wierenga PA, Vincken JP, Bastiaan-Net S, Weegels PL. Amylase trypsin inhibitors activation of toll-like receptor 4 revisited: The dominance of lipopolysaccharides contamination. Int J Biol Macromol 2025; 310:143378. [PMID: 40288707 DOI: 10.1016/j.ijbiomac.2025.143378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/12/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Amylase trypsin inhibitors (ATIs) potentially play a role in irritable bowel syndrome (IBS) and non-celiac wheat sensitivity (NCWS). These cereal-derived inhibitors are suspected to bind to the TLR4-MD2-CD14 complex and trigger intestinal pro-inflammatory responses, but confirmation through more extensive cell line studies is required. In this study, an amylase trypsin inhibitors enriched fraction (AEF) was prepared and characterized. Then, AEF binding potential to TLR4-MD2-CD14 was investigated using the human TLR4 reporter cell line HEK-BlueTM. The method took into account the presence of lipopolysaccharides (LPS) using Polymyxin B (PMB) to block LPS binding to TLR4. Proteinase K was also used to hydrolyze AEF proteins and eliminate their induced response. The cell line experiments showed that PMB treatment of AEF reduced the binding signal by 92 %. Complete hydrolysis of the protein by Proteinase K doubled the TLR4 activation signal and might indicate that protein-LPS complexation reduced LPS's ability to activate the TLR4-receptor. These finding underline the need for future work to consider the non-protein part in cell assays, especially the LPS bias. Altogether, these results indicated that LPS activates TLR4-MD2-CD14 and challenges ATIs' intestinal inflammation capacity contributing in irritable bowel syndrome and non-celiac wheat sensitivity.
Collapse
Affiliation(s)
- Dounia Krouch
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Gijs J C Vreeke
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Antoine H P America
- Wageningen Plant Research, Wageningen University & Research, The Netherlands, P.O. Box 16, 6700 AA Wageningen, the Netherlands
| | - Jurriaan J Mes
- Wageningen Food & Biobased Research, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Peter A Wierenga
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Jean-Paul Vincken
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands.
| | - Peter L Weegels
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands.
| |
Collapse
|
27
|
Pich K, Respekta-Długosz N, Kurowska P, Opydo M, Smolińska N, Dupont J, Rak A. Intelectin-1 promotes granulosa cells proliferation and modulates apoptosis via ERK1/2, AKT, and insulin receptor signaling pathways in Large White and Meishan pigs. Gen Comp Endocrinol 2025; 367:114722. [PMID: 40250633 DOI: 10.1016/j.ygcen.2025.114722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 04/20/2025]
Abstract
Maintaining the proper balance between granulosa cells (Gc) proliferation and apoptosis is crucial for folliculogenesis and female fertility. Our previous study showed expression of omentin-1 (intelectin-1, ITLN1) in the porcine ovarian follicles; however, its impact on Gc functions remains unknown. Therefore, this study aimed to determine the in vitro effects of ITLN1 on Gc proliferation and apoptosis in Large White (LW) and Meishan (MS) pigs. These breeds were chosen due to their distinct reproductive characteristics: MS pigs are known for maintaining a higher number of follicles during the follicular phase and exhibiting greater estradiol synthesis compared to LW pigs. Porcine Gc were incubated with ITLN1 (10-100 ng/mL) for 24-72 h, and the viability/proliferation (alamarBlue/BrdU assays), cell cycle progression (flow cytometry) and the gene and protein expression of proliferation/apoptotic markers (PCNA, cyclins A1, B2, D1, E1, caspases-3, -9, BCL-2, BAX, FAS, FADD, XIAP) (real-time PCR, western blotting) were assessed. Next, the effect of ITLN1 on the phosphorylation of several kinases (AKT, AMPK, ERK1/2, STAT3, PKA) and the gene and protein expression of the insulin receptor (INSR) were studied (real-time PCR, western blotting). Then, using pharmacological inhibitors of ERK1/2 (PD98059, 5 μM), AKT (LY294002, 10 μM) and INSR (1 μM), treated alone or with ITLN1 (S961, 50 ng/mL), we analyzed its involvement in the effects of ITLN1 on Gc proliferation/apoptosis. We demonstrated that ITLN1 had a mitogenic effect on Gc by enhancing cell cycle progression and modulating the levels of PCNA, cyclins and apoptotic factors via ERK1/2, AKT, and INSR, suggesting that ITLN1 is a newly identified regulator in ovarian folliculogenesis, regardless of the fatness degree of pigs.
Collapse
Affiliation(s)
- Karolina Pich
- Jagiellonian University in Krakow, Faculty of Biology, Institute of Zoology and Biomedical Research, Laboratory of Physiology and Toxicology of Reproduction, Poland; Jagiellonian University in Krakow, Doctoral School of Exact and Natural Sciences, Poland
| | - Natalia Respekta-Długosz
- Jagiellonian University in Krakow, Faculty of Biology, Institute of Zoology and Biomedical Research, Laboratory of Physiology and Toxicology of Reproduction, Poland; Jagiellonian University in Krakow, Doctoral School of Exact and Natural Sciences, Poland
| | - Patrycja Kurowska
- Jagiellonian University in Krakow, Faculty of Biology, Institute of Zoology and Biomedical Research, Laboratory of Physiology and Toxicology of Reproduction, Poland
| | - Małgorzata Opydo
- Jagiellonian University in Krakow, Faculty of Biology, Institute of Zoology and Biomedical Research, Laboratory of Experimental Hematology, Poland
| | - Nina Smolińska
- University of Warmia and Mazury in Olsztyn, Faculty of Biology and Biotechnology, Department of Animal Anatomy and Physiology, Poland
| | - Joëlle Dupont
- INRAE, Unité Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Agnieszka Rak
- Jagiellonian University in Krakow, Faculty of Biology, Institute of Zoology and Biomedical Research, Laboratory of Physiology and Toxicology of Reproduction, Poland.
| |
Collapse
|
28
|
Kumaraswamy A, Mannan R, Swaim OA, Rodansky E, Wang XM, Udager A, Mehra R, Li H, Morrissey C, Corey E, Haffner MC, Nelson PS, Chinnaiyan AM, Yates JA, Alumkal JJ. LSD1+8a is an RNA biomarker of neuroendocrine prostate cancer. Neoplasia 2025; 63:101151. [PMID: 40088674 PMCID: PMC11952868 DOI: 10.1016/j.neo.2025.101151] [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: 01/07/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND Lysine-specific demethylase 1 (LSD1) is a histone demethylase and regulator of differentiation, including in cancer. A neuronal-specific isoform of LSD1-LSD1+8a-has been shown to play a key role in promoting neuronal differentiation in the developing brain. We previously determined that LSD1+8a transcripts were detected in an aggressive subtype of prostate cancer harboring a neuronal program-neuroendocrine prostate cancer (NEPC)-but not in prostate adenocarcinomas harboring a glandular program. However, the number of samples examined was limited. METHODS Using a large collection of prostate cancer patient cell lines and patient-derived xenografts (PDXs), we measured LSD1+8a using quantitative polymerase chain reaction (qPCR), RNA in situ hybridization (RNA-ISH), and protein detection methods. We then validated our findings using an independent cohort of patient tumor samples. RESULTS LSD1+8a mRNA expression was detected in every NEPC cell line and PDX examined by qPCR and RNA-ISH but in none of the prostate adenocarcinomas. We validated the RNA-ISH results in patient tumors, confirming that LSD1+8a was expressed in all NEPC tumors but in none of the adenocarcinomas. Finally, we generated a rabbit monoclonal antibody specific to LSD1+8a protein and confirmed its specificity using normal neuronal tissue samples. However, LSD1+8a protein was not detectable in NEPC tumors-likely due to the substantially lower levels of LSD1+8a mRNA in NEPC tumors vs. normal neuronal tissues. CONCLUSIONS Measuring LSD1+8a mRNA is a sensitive and specific method for the diagnosis of NEPC, which is often challenging.
Collapse
Affiliation(s)
- Anbarasu Kumaraswamy
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Rahul Mannan
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Olivia A Swaim
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Eva Rodansky
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Xiao-Ming Wang
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Aaron Udager
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Rohit Mehra
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Hui Li
- RevMAb Biosciences, Burlingame, CA, USA
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Michael C Haffner
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Peter S Nelson
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, WA, USA; Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Arul M Chinnaiyan
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Urology, University of Michigan, Ann Arbor, MI, USA; Howard Hughes Medical Institute, Ann Arbor, MI, USA
| | - Joel A Yates
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Joshi J Alumkal
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Wang P, Sun X, He X, Kang D, Liu X, Liu D, Li A, Yang G, Lin Y, Li S, Wang Y, Wang Y. Glecirasib, a Potent and Selective Covalent KRAS G12C Inhibitor Exhibiting Synergism with Cetuximab or SHP2 Inhibitor JAB-3312. CANCER RESEARCH COMMUNICATIONS 2025; 5:792-803. [PMID: 40304209 PMCID: PMC12076188 DOI: 10.1158/2767-9764.crc-25-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/17/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
SIGNIFICANCE Glecirasib potently and selectively inhibits KRAS G12C and reduces ERK and AKT phosphorylation in KRAS G12C-mutant cancer cells, further inducing cell-cycle arrest and apoptosis. Glecirasib monotherapy leads to tumor regression in KRAS G12C-mutant animal models and shows synergistic effects with cetuximab or JAB-3312 (sitneprotafib).
Collapse
Affiliation(s)
- Peng Wang
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| | - Xin Sun
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| | - Xueting He
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| | - Di Kang
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| | - Xiaoyu Liu
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| | - Dan Liu
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| | - Amin Li
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| | - Guiqun Yang
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| | - Yiwei Lin
- Jacobio (US) Pharmaceuticals, Inc., Burlington, Massachusetts
| | - Sujing Li
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| | - Yinxiang Wang
- Jacobio Pharmaceuticals Group Co., Ltd., Beijing, China
| | - Yanping Wang
- Jacobio Pharmaceuticals Co., Ltd., Beijing, China
| |
Collapse
|
30
|
Justice JL, Greco TM, Hutton JE, Reed TJ, Mair ML, Botas J, Cristea IM. Multi-epitope immunocapture of huntingtin reveals striatum-selective molecular signatures. Mol Syst Biol 2025; 21:492-522. [PMID: 40169779 PMCID: PMC12048488 DOI: 10.1038/s44320-025-00096-3] [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: 09/07/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 04/03/2025] Open
Abstract
Huntington's disease (HD) is a debilitating neurodegenerative disorder affecting an individual's cognitive and motor abilities. HD is caused by a mutation in the huntingtin gene producing a toxic polyglutamine-expanded protein (mHTT) and leading to degeneration in the striatum and cortex. Yet, the molecular signatures that underlie tissue-specific vulnerabilities remain unclear. Here, we investigate this aspect by leveraging multi-epitope protein interaction assays, subcellular fractionation, thermal proteome profiling, and genetic modifier assays. The use of human cell, mouse, and fly models afforded capture of distinct subcellular pools of epitope-enriched and tissue-dependent interactions linked to dysregulated cellular pathways and disease relevance. We established an HTT association with nearly all subunits of the transcriptional regulatory Mediator complex (20/26), with preferential enrichment of MED15 in the tail domain. Using HD and KO models, we find HTT modulates the subcellular localization and assembly of the Mediator. We demonstrated striatal enriched and functional interactions with regulators of calcium homeostasis and chromatin remodeling, whose disease relevance was supported by HD fly genetic modifiers assays. Altogether, we offer insights into tissue- and localization-dependent (m)HTT functions and pathobiology.
Collapse
Affiliation(s)
- Joshua L Justice
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Todd M Greco
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Josiah E Hutton
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Tavis J Reed
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA
| | - Megan L Mair
- Jan and Dan Duncan Neurological Research Institute, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Juan Botas
- Jan and Dan Duncan Neurological Research Institute, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Washington Road, Princeton, NJ, 08544, USA.
| |
Collapse
|
31
|
Pich K, Pietroń K, Szlaga A, Billert M, Skrzypski M, Pawlicki P, Kotula-Balak M, Dupont J, Błasiak A, Rak A. Adipokines level in plasma, hypothalamus, ovaries and adipose tissue of rats with polycystic ovary syndrome. Reprod Biomed Online 2025; 50:104693. [PMID: 40199655 DOI: 10.1016/j.rbmo.2024.104693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 04/10/2025]
Abstract
RESEARCH QUESTION Do the levels of adipokines (adiponectin, apelin, chemerin and vaspin) in plasma, hypothalamus, ovaries and periovarian adipose tissue differ during polycystic ovarian syndrome (PCOS)? DESIGN The PCOS was induced in rats by oral administration of non-steroidal aromatase inhibitor letrozole. To determine the plasma levels of adiponectin, apelin, chemerin and vaspin enzyme-linked immunosorbent assays were carried out. To assess the expression (gene and protein) and immunolocalization of these adipokines and their receptors, namely Adipor1 and Adipor2 for adiponectin, Aplnr for apelin, Ccrl2, Cmklr1 and Gpr1 for chemerin and Grp78 for vaspin in the hypothalamus, real-time polymerase chain reaction, and Western blot and immunohistochemistry were used to analyse ovaries and periovarian adipose tissue respectively. RESULTS In PCOS, the plasma level of adiponectin decreased (P = 0.0003), whereas apelin, chemerin and vaspin increased (P ≤ 0.0479). Moreover, PCOS modulates the expression of adipokines and their receptors in the hypothalamus, ovaries and periovarian adipose tissue compared with healthy rats (P ≤ 0.487). CONCLUSIONS A strong relationship was found between PCOS and adipokines, which suggests that adipokines may be a biomarker of PCOS.
Collapse
Affiliation(s)
- Karolina Pich
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Kraków, Poland; Doctoral School of Exact and Natural Sciences, Jagiellonian University in Krakow, Poland
| | - Klaudia Pietroń
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Kraków, Poland
| | - Agata Szlaga
- Departament of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| | - Maria Billert
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, Poznań, Poland
| | - Marek Skrzypski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, Poznań, Poland
| | - Piotr Pawlicki
- Center of Experimental and Innovative Medicine, University of Agriculture in Kraków, Kraków, Poland
| | - Małgorzata Kotula-Balak
- Department of Animal Anatomy and Preclinical Sciences, University Centre of Veterinary Medicine JU-UA, University of Agriculture in Kraków, Kraków Poland
| | - Joëlle Dupont
- INRAE, Physiologie de la Reproduction et des Comportements, Nouzilly, France
| | - Anna Błasiak
- Departament of Neurophysiology and Chronobiology, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Poland
| | - Agnieszka Rak
- Laboratory of Physiology and Toxicology of Reproduction, Institute of Zoology and Biomedical Research, Jagiellonian University in Kraków, Kraków, Poland.
| |
Collapse
|
32
|
Zhang HY, Fan ZL, Wang C, Li JY, Feng HG, Wang XY, Wang TY. Improved recombinant protein expression using the 5'-untranslated region in Chinese hamster ovary cells. Int J Biol Macromol 2025; 309:142822. [PMID: 40187442 DOI: 10.1016/j.ijbiomac.2025.142822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Chinese hamster ovary (CHO) cells are major expression platforms for the transient production of recombinant therapeutic proteins (RTPs). Most improvement strategies have focused on promoting transcriptional expression in CHO cells. However, methods for promoting the yield of RTPs through translational regulation remain unclear. In this study, we investigated characteristics of the 5'-untranslated region (UTR) that influence recombinant protein expression in CHO cells and identified sequences that have positive effects on protein expression using ribosome sequencing. Some elements and characteristics of 5'-UTR differentially affected the translation of the main open reading frame and increased recombinant protein expression by 1.5-fold in CHO cells. The findings may help relieve the bottleneck of the yield of RTPs on translation enhancement.
Collapse
Affiliation(s)
- Huan-Yu Zhang
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang 453000, China; Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China; International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453000, China
| | - Zhen-Lin Fan
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453000, China
| | - Chong Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453000, China
| | - Jia-Yue Li
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang 453000, China; International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453000, China
| | - Hui-Gen Feng
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453000, China; College of Life Science and Technology, North Henan Medical University, Xinxiang 453003, Henan, China.
| | - Xiao-Yin Wang
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang 453000, China; International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453000, China.
| | - Tian-Yun Wang
- Department of Biochemistry and Molecular Biology, Xinxiang Medical University, Xinxiang 453000, China; International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453000, China.
| |
Collapse
|
33
|
Thompson A, May MR, Hopkins BR, Riedl N, Barmina O, Liebeskind BJ, Zhao L, Begun D, Kopp A. Quantifying Transcriptome Turnover on Phylogenies by Modeling Gene Expression as a Binary Trait. Mol Biol Evol 2025; 42:msaf106. [PMID: 40423579 PMCID: PMC12108096 DOI: 10.1093/molbev/msaf106] [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: 10/15/2024] [Revised: 03/12/2025] [Accepted: 04/09/2025] [Indexed: 05/28/2025] Open
Abstract
Changes in gene expression are a key driver of phenotypic evolution, leading to a persistent interest in the evolution of transcriptomes. Traditionally, gene expression is modeled as a continuous trait, leaving qualitative transitions largely unexplored. In this paper, we detail the development of new Bayesian inference techniques to study the evolutionary turnover of organ-specific transcriptomes, which we define as instances where orthologous genes gain or lose expression in a particular organ. To test these techniques, we analyze the transcriptomes of 2 male reproductive organs, testes and accessory glands, across 11 species of the Drosophila melanogaster species group. We first discretize gene expression states by estimating the probability that each gene is expressed in each organ and species. We then define a phylogenetic model of correlated transcriptome evolution in 2 or more organs and fit it to the expression state data. Inferences under this model imply that many genes have gained and lost expression in each organ, and that the 2 organs experienced accelerated transcriptome turnover on different branches of the Drosophila phylogeny.
Collapse
Affiliation(s)
- Ammon Thompson
- Department of Evolution and Ecology, University of California, Davis, CA, USA
| | - Michael R May
- Department of Evolution and Ecology, University of California, Davis, CA, USA
| | - Ben R Hopkins
- Department of Evolution and Ecology, University of California, Davis, CA, USA
| | - Nerisa Riedl
- Department of Evolution and Ecology, University of California, Davis, CA, USA
| | - Olga Barmina
- Department of Evolution and Ecology, University of California, Davis, CA, USA
| | - Benjamin J Liebeskind
- Center for Systems and Synthetic Biology, Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Li Zhao
- Department of Evolution and Ecology, University of California, Davis, CA, USA
- Laboratory of Evolutionary Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - David Begun
- Department of Evolution and Ecology, University of California, Davis, CA, USA
| | - Artyom Kopp
- Department of Evolution and Ecology, University of California, Davis, CA, USA
| |
Collapse
|
34
|
Gao Q, Zhang Z, Fu R, Zhu C, Yuwen W, Xu R, Duan Z, Fan D. Expression, optimization and biological activity analysis of recombinant type XII collagen in Pichia pastoris. Int J Biol Macromol 2025; 311:143720. [PMID: 40316097 DOI: 10.1016/j.ijbiomac.2025.143720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/02/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
Collagen XII (COL12A1) is a type of FACIT collagen that plays an important role in the extracellular matrix structuring, participating in the regulation of collagen fiber size, and serves as a link between different components of the extracellular matrix. However, it is still unclear whether exogenous administration of collagen XII has a direct regulatory effect. In this study, we successfully produced recombinant human XII-type collagen (rh12C) through genetic engineering approach, which is composed of different functional domains. A Pichia pastoris host cell strain was constructed based on the intracellular translation regulatory mechanism of collagen, achieving a maximum yield of 4.89 g/L. After purification and structural characterization of the protein, its potential biological efficacy was evaluated through in vitro cell experiments.
Collapse
Affiliation(s)
- Qiao Gao
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering and Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China; Xi'an Synthetic Biology Technology and Biomaterials International Science and Technology Cooperation Base, School of Chemical Engineering, Northwest University, Xi'an 710127, China
| | - Zhuo Zhang
- Plastic and Cosmetic Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, China
| | - Rongzhan Fu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering and Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China; Xi'an Synthetic Biology Technology and Biomaterials International Science and Technology Cooperation Base, School of Chemical Engineering, Northwest University, Xi'an 710127, China
| | - Chenhui Zhu
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering and Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China; Xi'an Synthetic Biology Technology and Biomaterials International Science and Technology Cooperation Base, School of Chemical Engineering, Northwest University, Xi'an 710127, China
| | - Weigang Yuwen
- Shaanxi Giant Biotechnology Co., Ltd, Xi'an 710065, Shaanxi, China
| | - Ru Xu
- Shaanxi Giant Biotechnology Co., Ltd, Xi'an 710065, Shaanxi, China
| | - Zhiguang Duan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering and Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China; Xi'an Synthetic Biology Technology and Biomaterials International Science and Technology Cooperation Base, School of Chemical Engineering, Northwest University, Xi'an 710127, China.
| | - Daidi Fan
- Engineering Research Center of Western Resource Innovation Medicine Green Manufacturing, Ministry of Education, School of Chemical Engineering, Northwest University, Xi'an 710069, China; Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering and Biotech. & Biomed. Research Institute, Northwest University, Xi'an 710069, China; Xi'an Synthetic Biology Technology and Biomaterials International Science and Technology Cooperation Base, School of Chemical Engineering, Northwest University, Xi'an 710127, China.
| |
Collapse
|
35
|
Northoff BH, Herbst A, Wenk C, Weindl L, Gäbel G, Brezski A, Zarnack K, Küpper A, Dimmeler S, Moretti A, Laugwitz KL, Engelhardt S, Maegdefessel L, Boon RA, Doppler S, Dreßen M, Lahm H, Lange R, Krane M, Krohn K, Kohlmaier A, Holdt LM, Teupser D. Circular RNAs increase during vascular cell differentiation and are biomarkers for vascular disease. Cardiovasc Res 2025; 121:405-423. [PMID: 39901821 PMCID: PMC12038242 DOI: 10.1093/cvr/cvaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 07/31/2024] [Accepted: 12/12/2024] [Indexed: 02/05/2025] Open
Abstract
AIMS The role of circular RNAs (circRNAs) and their regulation in health and disease are poorly understood. Here, we systematically investigated the temporally resolved transcriptomic expression of circRNAs during differentiation of human induced pluripotent stem cells (iPSCs) into vascular endothelial cells (ECs) and smooth muscle cells (SMCs) and explored their potential as biomarkers for human vascular disease. METHODS AND RESULTS Using high-throughput RNA sequencing and a de novo circRNA detection pipeline, we quantified the daily levels of 31 369 circRNAs in a 2-week differentiation trajectory from human stem cells to proliferating mesoderm progenitors to quiescent, differentiated EC and SMC. We detected a significant global increase in RNA circularization, with 397 and 214 circRNAs up-regulated greater than two-fold (adjusted P < 0.05) in mature EC and SMC, compared with undifferentiated progenitor cells. This global increase in circRNAs was associated with up-regulation of host genes and their promoters and a parallel down-regulation of splicing factors. Underlying this switch, the proliferation-regulating transcription factor MYC decreased as vascular cells matured, and inhibition of MYC led to down-regulation of splicing factors such as SRSF1 and SRSF2 and changes in vascular circRNA levels. Examining the identified circRNAs in arterial tissue samples and in peripheral blood mononuclear cells (PBMCs) from patients, we found that circRNA levels decreased in atherosclerotic disease, in contrast to their increase during iPSC maturation into EC and SMC. Using machine learning, we determined that a set of circRNAs derived from COL4A1, COL4A2, HSPG2, and YPEL2 discriminated atherosclerotic from healthy tissue with an area under the receiver operating characteristic curve (AUC) of 0.79. circRNAs from HSPG2 and YPEL2 in blood PBMC samples detected atherosclerosis with an AUC of 0.73. CONCLUSION Time-resolved transcriptional profiling of linear and circRNA species revealed that circRNAs provide granular molecular information for disease profiling. The identified circRNAs may serve as blood biomarkers for atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Bernd H Northoff
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Andreas Herbst
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Catharina Wenk
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Lena Weindl
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Gabor Gäbel
- Department of Vascular Medicine, HELIOS Klinikum Krefeld, Krefeld, Germany
| | - Andre Brezski
- Buchmann Institute for Molecular Life Sciences (BMLS), Faculty of Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Kathi Zarnack
- Buchmann Institute for Molecular Life Sciences (BMLS), Faculty of Biological Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Alina Küpper
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Stefanie Dimmeler
- Institute of Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Alessandra Moretti
- Department of Internal Medicine I, Cardiology, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Karl-Ludwig Laugwitz
- Department of Internal Medicine I, Cardiology, Klinikum rechts der Isar, School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Stefan Engelhardt
- Institute of Pharmacology and Toxicology, Technical University of Munich (TUM), Munich, Germany
| | - Lars Maegdefessel
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany
| | - Reinier A Boon
- Institute of Cardiovascular Regeneration, Centre of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Stefanie Doppler
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University Munich, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University Munich, Munich, Germany
| | - Martina Dreßen
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University Munich, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University Munich, Munich, Germany
| | - Harald Lahm
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University Munich, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University Munich, Munich, Germany
| | - Rüdiger Lange
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University Munich, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Markus Krane
- Department of Cardiovascular Surgery, German Heart Center Munich, Technical University Munich, Munich, Germany
- Institute for Translational Cardiac Surgery (INSURE), German Heart Center Munich, Technical University Munich, Munich, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
- Division of Cardiac Surgery, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Knut Krohn
- Core Unit DNA Technologies, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Alexander Kohlmaier
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Lesca M Holdt
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| | - Daniel Teupser
- Institute of Laboratory Medicine, LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377 Munich, Germany
| |
Collapse
|
36
|
Wagner TM, Torres-Puig S, Yimthin T, Irobalieva RN, Heller M, Kaessmeyer S, Démoulins T, Jores J. Extracellular vesicles of minimalistic Mollicutes as mediators of immune modulation and horizontal gene transfer. Commun Biol 2025; 8:674. [PMID: 40301684 PMCID: PMC12041197 DOI: 10.1038/s42003-025-08099-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/16/2025] [Indexed: 05/01/2025] Open
Abstract
Extracellular vesicles (EVs) are central components of bacterial secretomes, including the small, cell wall-less Mollicutes. Although EV release in Mollicutes has been reported, EV proteomic composition and function have not been explored yet. We developed a protocol for isolating EVs of the pathogens Mycoplasma mycoides subsp. capri (Mmc) and Mycoplasma (Mycoplasmopsis) bovis and examined their functionality. Proteomic analysis demonstrated that EVs mirror the proteome of the EV-producing bacteria. EVs exhibited nuclease activity, effectively digesting both circular and linear DNA. Notably, M. bovis EVs elicited immune responses in bovine primary blood cells, like those induced by live M. bovis. Our findings reveal that EVs can carry plasmids and enable their horizontal transfer, known as vesiduction. Specifically, the natural plasmid pKMK1, with an unknown transmission route, was detected in EVs of Mmc 152/93 and the tetM-containing pIVB08 plasmid was associated with EVs released by an Mmc GM12 strain carrying this plasmid. pIVB08 could be transferred via homo- and heterologous vesiduction to Mmc, M. capricolum subsp. capricolum and M. leachii. Vesiduction was impeded by membrane disruption but resisted DNase and Proteinase K treatment, suggesting that EVs protect their cargo. These findings enhance our understanding of Mollicutes EVs, particularly in host interactions and horizontal gene transfer.
Collapse
Affiliation(s)
- Theresa Maria Wagner
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland.
| | - Sergi Torres-Puig
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland
| | - Thatcha Yimthin
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, Bern, Switzerland
| | - Rossitza N Irobalieva
- Division of Veterinary Anatomy, Department of Clinical Research and Veterinary Public Health, University of Bern, Bern, Switzerland
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Sabine Kaessmeyer
- Division of Veterinary Anatomy, Department of Clinical Research and Veterinary Public Health, University of Bern, Bern, Switzerland
| | - Thomas Démoulins
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland
| | - Jörg Jores
- Institute of Veterinary Bacteriology, Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty - University of Bern, Bern, Switzerland.
- Multidisciplinary Center for Infectious Diseases (MCID), University of Bern, Bern, Switzerland.
| |
Collapse
|
37
|
Bhuiyan T, Arecco N, Mendoza Sanchez PK, Kim J, Schwan C, Weyrauch S, Nizamuddin S, Prunotto A, Tekman M, Biniossek ML, Knapp B, Koidl S, Drepper F, Huesgen PF, Grosse R, Hugel T, Arnold SJ. TAF2 condensation in nuclear speckles links basal transcription factor TFIID to RNA splicing factors. Cell Rep 2025; 44:115616. [PMID: 40287942 DOI: 10.1016/j.celrep.2025.115616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 11/22/2024] [Accepted: 04/01/2025] [Indexed: 04/29/2025] Open
Abstract
TFIID is an essential basal transcription factor, crucial for RNA polymerase II (pol II) promoter recognition and transcription initiation. The TFIID complex consists of the TATA binding protein (TBP) and 13 TBP-associated factors (TAFs) that contain intrinsically disordered regions (IDRs) with currently unknown functions. Here, we show that a conserved IDR drives TAF2 to nuclear speckle condensates independently of other TFIID subunits. Quantitative mass spectrometry analyses reveal TAF2 proximity to RNA splicing factors including specific interactions of the TAF2 IDR with SRRM2 in nuclear speckles. Deleting the IDR from TAF2 does not majorly impact global gene expression but results in changes of alternative splicing events. Further, genome-wide binding analyses suggest that the TAF2 IDR impedes TAF2 promoter association by guiding TAF2 to nuclear speckles. This study demonstrates that an IDR within the large multiprotein complex TFIID controls nuclear compartmentalization and thus links distinct molecular processes, namely transcription initiation and RNA splicing.
Collapse
Affiliation(s)
- Tanja Bhuiyan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstrasse 25, 79104 Freiburg, Germany; Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, Breisacher Strasse 66, 79106 Freiburg, Germany.
| | - Niccolò Arecco
- Genome Biology Unit, Centre for Genomic Regulation (CRG), 08003 Barcelona, Spain
| | - Paulina Karen Mendoza Sanchez
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, Breisacher Strasse 66, 79106 Freiburg, Germany; German Cancer Consortium (DKTK) Partner Site Freiburg, 79106 Freiburg, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Juhyeong Kim
- Institute of Physical Chemistry, University of Freiburg, Albertstrasse 21, 79104 Freiburg, Germany
| | - Carsten Schwan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstrasse 25, 79104 Freiburg, Germany
| | - Sophie Weyrauch
- Institute of Physical Chemistry, University of Freiburg, Albertstrasse 21, 79104 Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Alberstrasse 19A, 79104 Freiburg, Germany
| | - Sheikh Nizamuddin
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, Breisacher Strasse 66, 79106 Freiburg, Germany; German Cancer Consortium (DKTK) Partner Site Freiburg, 79106 Freiburg, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Andrea Prunotto
- Datenintegrationszentrum, Medical Center-University of Freiburg, Faculty of Medicine, Georges-Köhler-Allee 302, 79110 Freiburg, Germany
| | - Mehmet Tekman
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstrasse 25, 79104 Freiburg, Germany
| | - Martin L Biniossek
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Stefan-Meier-Strasse 17, 79104 Freiburg, Germany
| | - Bettina Knapp
- Institute for Biology II, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany
| | - Stefanie Koidl
- Department of Urology, Medical Center-University of Freiburg, Faculty of Medicine, Breisacher Strasse 66, 79106 Freiburg, Germany; German Cancer Consortium (DKTK) Partner Site Freiburg, 79106 Freiburg, Germany; German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Friedel Drepper
- Institute for Biology II, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany
| | - Pitter F Huesgen
- Institute for Biology II, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany; BIOSS and CIBSS Signalling Research Centres, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
| | - Robert Grosse
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstrasse 25, 79104 Freiburg, Germany; BIOSS and CIBSS Signalling Research Centres, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
| | - Thorsten Hugel
- Institute of Physical Chemistry, University of Freiburg, Albertstrasse 21, 79104 Freiburg, Germany; BIOSS and CIBSS Signalling Research Centres, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
| | - Sebastian J Arnold
- Institute of Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Freiburg, Albertstrasse 25, 79104 Freiburg, Germany; BIOSS and CIBSS Signalling Research Centres, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany.
| |
Collapse
|
38
|
Leduc A, Zheng S, Saxena P, Slavov N. Impact of protein degradation and cell growth on mammalian proteomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637566. [PMID: 39990504 PMCID: PMC11844506 DOI: 10.1101/2025.02.10.637566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cellular protein concentrations are controlled by rates of synthesis and clearance, the lat-ter including protein degradation and dilution due to growth. Thus, cell growth rate may influence the mechanisms controlling variation in protein concentrations. To quantify this influence, we analyzed the growth-dependent effects of protein degradation within a cell type (between activated and resting human B-cells), across human cell types and mouse tissues. This analysis benefited from deep and accurate quantification of over 12,000 proteins across four primary tissues using plexDIA. The results indicate that growth-dependent dilution can account for 40 % of protein concentration changes across conditions. Furthermore, we find that the variation in protein degradation rates is sufficient to account for up to 50 % of the variation in concentrations within slowly growing cells as contrasted with 7 % in growing cells. Remarkably, degradation rates differ significantly between proteoforms encoded by the same gene and arising from alternative splicing or alternate RNA decoding. These proteoform-specific degradation rates substantially determine the proteoform abundance, especially in the brain. Thus, our model and data unify previous observations with our new results and demonstrate substantially larger than previously appreciated contributions of protein degradation to protein variation at slow growth, both across proteoforms and tissue types.
Collapse
|
39
|
Xu M, Zhang QT, Zhou L, Cai YW, Liu H, Zhao QL, Tian JH, Huang YK, Ren P, Huang X. Ferulic acid in Chaihu Shugan San modulates depression-like behavior, endothelial and gastrointestinal dysfunction in rats via the Ghrl-Edn1/Mecp2/P-mTOR/VEGFA pathway: A multi-omics study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119624. [PMID: 40127829 DOI: 10.1016/j.jep.2025.119624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/16/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In a global context of escalating multimorbidity, characterized by the co-occurrence of major depressive disorder (MDD), endothelial dysfunction (ED), and gastrointestinal dysregulation (GD), the quest for effective treatments has become paramount. Central to these interconnected conditions is oxidative stress (OS), a pivotal factor that has been extensively studied yet remains inadequately addressed. This study introduces Chaihu-Shugan-San (CSS) and its absorbed component ferulic acid (FA), a potent antioxidant derived from medicinal plants, as a novel therapeutic approach with the unique ability to counter the multifaceted effects of acute forced swimming (AFS)-induced depression, ED, and GD. Unlike traditional single-disease-focused studies, our research explores the synergistic effects of CSS and FA across these interrelated disorders, offering a groundbreaking perspective. AIM OF THE STUDY This study aims to evaluate CSS and FA in treating depression-related multimorbidity triggered by AFS and to uncover the shared underlying mechanisms of FA. MATERIALS AND METHODS A depression-like model in rats was induced by AFS, and an OS model was established in endothelial cells (ECs) through hydrogen peroxide treatment. We investigated the effects of CSS and FA on MDD, ED, and GD in rats and OS levels in ECs. Our assessments included hematoxylin and eosin (HE) staining, biochemical assays, and behavioral studies. We conducted an integrated analysis of transcriptomics, proteomics, and phosphoproteomics data to elucidate the underlying mechanisms. The identification of relevant targets was confirmed through Western blotting (WB), real-time quantitative polymerase chain reaction (RT-qPCR), molecular docking studies, and an extensive literature review. RESULTS Our findings indicate that CSS and FA not only significantly mitigate AFS-induced abnormalities in the open field test (OFT), forced swim test (FST), and related behaviors such as gastric emptying and intestinal transit in rats but also ameliorate depression, ED, GD, inflammation and OS-related biomarker levels, alongside HE staining in gastric sinus and aorta slices. The study also highlights that FA can influence OS and endothelial function in ECs. Moreover, a combined multi-omics analysis unveiled several OS-related pathways, including the mTOR and p53 signaling pathways. Our research elucidates that the Ghrl-Edn1/Mecp2/P-mTOR/Vegfa-associated OS signaling pathway is pivotal in countering AFS-induced multimorbidity, expanding beyond the conventional disease-specific focus. CONCLUSIONS This pioneering study underscores capability of CSS and FA to tackle AFS-induced multimorbidity concurrently and intricately details FA's antioxidative mechanisms within ECs. The insights gleaned offer a novel perspective on FA's role in multimorbidity regulation and its potential to modulate OS, especially in the complex environment of ECs. Given the urgent global health challenges, our research positions FA as a promising therapeutic contender, advocating for a paradigm shift in multimorbidity management.
Collapse
Affiliation(s)
- Min Xu
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qian-Tao Zhang
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Zhou
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ya-Wen Cai
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Liu
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiu-Long Zhao
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Hua Tian
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun-Ke Huang
- Women's Hospital, Zhejiang University School of Medicine, Gynecology Department, Zhejiang, China
| | - Ping Ren
- Department of Geriatrics, Jiangsu Province Hospital of TCM, Nanjing, China
| | - Xi Huang
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
40
|
Krishna CK, Das H, Hohnen L, Schliebs W, Oeljeklaus S, Warscheid B, Kalel VC, Erdmann R. High-confidence glycosomal membrane protein inventory unveils trypanosomal peroxin PEX15. Cell Rep 2025; 44:115614. [PMID: 40286272 DOI: 10.1016/j.celrep.2025.115614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/11/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Trypanosomatid parasite infections cause Chagas disease, human African trypanosomiasis, and leishmaniasis, affecting over 12 million people worldwide. Glycosomes, the peroxisome-related organelles of trypanosomes, are essential for survival, making their metabolic functions and biogenesis mediated by peroxins (PEXs) suitable drug targets. We report a comprehensive protein inventory of glycosomal membranes, defined through advanced subcellular membrane protein profiling combined with quantitative mass spectrometry and including 28 high-confidence glycosomal membrane proteins. We validate four previously unknown glycosomal membrane proteins, including a tail-anchored protein, which we show to be the long-sought Trypanosoma PEX15. Despite low sequence similarity, Trypanosoma PEX15 exhibits structural and topological similarities with its yeast and human counterparts, and it is essential for glycosome biogenesis and parasite survival. Considering the low degree of conservation with its human counterpart, PEX15 is a promising target for drug development. This inventory is an important resource for characterizing glycosome biology and therapeutic development.
Collapse
Affiliation(s)
- Chethan K Krishna
- Department of Systems Biochemistry, Institute of Biochemistry and Pathobiochemistry, Faculty of Medicine, Ruhr University Bochum, Bochum, Germany
| | - Hirak Das
- Biochemistry II, Theodor-Boveri-Institute, Biocenter, Faculty of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany
| | - Lisa Hohnen
- Department of Systems Biochemistry, Institute of Biochemistry and Pathobiochemistry, Faculty of Medicine, Ruhr University Bochum, Bochum, Germany
| | - Wolfgang Schliebs
- Department of Systems Biochemistry, Institute of Biochemistry and Pathobiochemistry, Faculty of Medicine, Ruhr University Bochum, Bochum, Germany
| | - Silke Oeljeklaus
- Biochemistry II, Theodor-Boveri-Institute, Biocenter, Faculty of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany
| | - Bettina Warscheid
- Biochemistry II, Theodor-Boveri-Institute, Biocenter, Faculty of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany.
| | - Vishal C Kalel
- Department of Systems Biochemistry, Institute of Biochemistry and Pathobiochemistry, Faculty of Medicine, Ruhr University Bochum, Bochum, Germany.
| | - Ralf Erdmann
- Department of Systems Biochemistry, Institute of Biochemistry and Pathobiochemistry, Faculty of Medicine, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
41
|
Brunchault MR, Hesse AM, Schaeffer J, Fröhlich A, Saintpierre A, Decourt C, Combes F, Nawabi H, Couté Y, Belin S. Proteomics-based characterization of ribosome heterogeneity in adult mouse organs. Cell Mol Life Sci 2025; 82:175. [PMID: 40272563 PMCID: PMC12022211 DOI: 10.1007/s00018-025-05708-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 03/25/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
The translation process, leading to protein synthesis from mRNA, has been long thought to be invariable in all cellular organisms. Increasing evidence shows that it is finely regulated by variable features of the translation machinery. Notably, ribosomes, the functional units of protein synthesis, are suggested to display variations in their composition, depending on the developmental stage, cell type or physio-pathological context, thus hinting a new level of actionable regulation of gene expression. Yet, a comprehensive map of the heterogeneity of ribosome composition in ribosomal proteins (RPs) in different organs and tissues is not available. In this work, we explored tissue-specific ribosome heterogeneity using mass spectrometry-based quantitative proteomic characterization of ribosomal fractions purified from 14 adult mouse organs and tissues. We performed crossed clustering and statistical analyses of RP composition to highlight stable, variable and tissue-specific RPs across organs and tissues. Focusing on specific RPs, we validated their varying abundances using a targeted proteomic approach and western blot analyses, providing further insights into the tissue-specific ribosome RP signature. Finally, we investigated the origin of RP variations in ribosome fraction of the different tissues, by comparing RP relative amounts in our ribosomal proteomic dataset with their corresponding transcript abundances in three independent transcriptomic datasets. Interestingly, we found that, in some tissues, the RP abundance in purified ribosomes does not always correlate with the corresponding RP transcript level, arguing for a translational regulation of RP expression, and/or a regulated incorporation of RPs into ribosomes. Altogether, our data support the notion of a tissue-specific RP signature of ribosomes, which opens avenues to study how specific ribosomal composition provides an additional level of regulation to control gene expression in different tissues and organs.
Collapse
Affiliation(s)
- Marie R Brunchault
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Anne-Marie Hesse
- Univ. Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, 38000, Grenoble, France
| | - Julia Schaeffer
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
- IBDM, CNRS, UMR 7288, Aix-Marseille Université, Marseille, France
| | - Albrecht Fröhlich
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Ana Saintpierre
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Charlotte Decourt
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Florence Combes
- Univ. Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, 38000, Grenoble, France
| | - Homaira Nawabi
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France
| | - Yohann Couté
- Univ. Grenoble Alpes, INSERM, CEA, UA13 BGE, CNRS, CEA, FR2048, 38000, Grenoble, France.
| | - Stephane Belin
- Univ. Grenoble Alpes, Inserm, U1216, Grenoble Institut Neurosciences, 38000, Grenoble, France.
| |
Collapse
|
42
|
Mariano NC, Marotti JD, Chen Y, Karakyriakou B, Salgado R, Christensen BC, Miller TW, Kettenbach AN. Quantitative proteomics analysis of triple-negative breast cancers. NPJ Precis Oncol 2025; 9:117. [PMID: 40269124 PMCID: PMC12019170 DOI: 10.1038/s41698-025-00907-8] [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: 09/11/2024] [Accepted: 04/05/2025] [Indexed: 04/25/2025] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for approximately 15% of all Breast Cancer (BC) cases with poorer prognosis and clinical outcomes compared to other BC subtypes due to greater tumor heterogeneity and few therapeutically targetable oncogenic drivers. To reveal actionable pathways for anti-cancer treatment, we use a proteomic approach to quantitatively compare the abundances of 6306 proteins across 55 formalin-fixed and paraffin-embedded (FFPE) TNBC tumors. We identified four major TNBC clusters by unsupervised clustering analysis of protein abundances. Analyses of clinicopathological characteristics revealed associations between the proteomic profiles and clinical phenotypes exhibited by each subtype. We validate the findings by inferring immune and stromal cell type composition from genome-wide DNA methylation profiles. Finally, quantitative proteomics on TNBC cell lines was conducted to identify in vitro models for each subtype. Collectively, our data provide subtype-specific insights into molecular drivers, clinicopathological phenotypes, tumor microenvironment (TME) compositions, and potential pharmacologic vulnerabilities for further investigations.
Collapse
Affiliation(s)
| | - Jonathan D Marotti
- Department of Pathology and Laboratory Medicine, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
| | | | | | - Roberto Salgado
- Department of Pathology, GZA-ZNA Hospitals, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Brock C Christensen
- Department of Pathology and Laboratory Medicine, Lebanon, NH, USA
- Dartmouth Cancer Center, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Lebanon, NH, USA
- Department of Epidemiology, Lebanon, NH, USA
- Department of Community and Family Medicine, Lebanon, NH, USA
| | - Todd W Miller
- Dartmouth Cancer Center, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Lebanon, NH, USA
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Hanover, NH, USA.
- Dartmouth Cancer Center, Lebanon, NH, USA.
| |
Collapse
|
43
|
Zheng P, Audain E, Webel H, Dai C, Klein J, Hitz MP, Sachsenberg T, Bai M, Perez-Riverol Y. Ibaqpy: A scalable Python package for baseline quantification in proteomics leveraging SDRF metadata. J Proteomics 2025; 317:105440. [PMID: 40268243 DOI: 10.1016/j.jprot.2025.105440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Intensity-based absolute quantification (iBAQ) is essential in proteomics as it allows for the assessment of a protein's absolute abundance in various samples or conditions. However, the computation of these values for increasingly large-scale and high-throughput experiments, such as those using DIA, TMT, or LFQ workflows, poses significant challenges in scalability and reproducibility. Here, we present ibaqpy (https://github.com/bigbio/ibaqpy), a Python package designed to compute iBAQ values efficiently for experiments of any scale. Ibaqpy leverages the Sample and Data Relationship Format (SDRF) metadata standard to incorporate experimental metadata into the quantification workflow. This allows for automatic normalization and batch correction while accounting for key aspects of the experimental design, such as technical and biological replicates, fractionation strategies, and sample conditions. Designed for large-scale proteomics datasets, ibaqpy can also recompute iBAQ values for existing experiments when an SDRF is available. We showcased ibaqpy's capabilities by reanalyzing 17 public proteomics datasets from ProteomeXchange, covering HeLa cell lines with 4921 samples and 5766 MS runs, quantifying a total of 11,014 proteins. In our reanalysis, ibaqpy is a key component in automating reproducible quantification, reducing manual effort and making quantitative proteomics more accessible while supporting FAIR principles for data reuse. SIGNIFICANCE: Proteomics studies often rely on intensity-based absolute quantification (iBAQ) to assess protein abundance across various biological conditions. Despite its widespread use, computing iBAQ values at scale remains challenging due to the increasing complexity and volume of proteomics experiments. Existing tools frequently lack metadata integration, limiting their ability to handle experimental design intricacies such as replicates, fractions, and batch effects. Our work introduces ibaqpy, a scalable Python package that leverages the Sample and Data Relationship Format (SDRF) to compute iBAQ values efficiently while incorporating critical experimental metadata. By enabling automated normalization and batch correction, ibaqpy ensures reproducible and comparable quantification across large-scale datasets. We validated the utility of ibaqpy through the reanalysis of 17 public HeLa datasets, comprising over 200 million peptide features and quantifying 11,000 proteins across thousands of samples. This comprehensive reanalysis highlights the robustness and scalability of ibaqpy, making it an essential tool for researchers conducting large-scale proteomics experiments. Moreover, by promoting FAIR principles for data reuse and interoperability, ibaqpy offers a transformative approach to baseline protein quantification, supporting reproducible research and data integration within the proteomics community.
Collapse
Affiliation(s)
- Ping Zheng
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing, China
| | - Enrique Audain
- Institute of Medical Genetics, University Medicine Oldenburg, Carl von Ossietzky University, Oldenburg, Germany
| | - Henry Webel
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - Chengxin Dai
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Life Omics, Beijing, China
| | - Joshua Klein
- Program for Bioinformatics, Boston University, Boston, USA
| | - Marc-Phillip Hitz
- Institute of Medical Genetics, University Medicine Oldenburg, Carl von Ossietzky University, Oldenburg, Germany
| | - Timo Sachsenberg
- Department of Computer Science, Applied Bioinformatics, University of Tübingen, Tübingen, Germany; Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany
| | - Mingze Bai
- Chongqing Key Laboratory of Big Data for Bio Intelligence, Chongqing University of Posts and Telecommunications, Chongqing, China.
| | - Yasset Perez-Riverol
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, UK.
| |
Collapse
|
44
|
Landgraf A, Okada J, Horton M, Liu L, Solomon S, Qiu Y, Kurland IJ, Sidoli S, Pessin JE, Shinoda K. Widespread discordance between mRNA expression, protein abundance and de novo lipogenesis activity in hepatocytes during the fed-starvation transition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.15.649020. [PMID: 40376090 PMCID: PMC12080948 DOI: 10.1101/2025.04.15.649020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
The mammalian liver plays a critical role in maintaining metabolic homeostasis during fasting and feeding. Liver function is further shaped by sex dimorphism and zonation of hepatocytes. To explore how these factors interact, we performed deep RNA-sequencing and label-free proteomics on periportal and pericentral hepatocytes isolated from male and female mice under fed and starved conditions. We developed a classification system to assess protein-mRNA relationship and found that gene products (mRNA or protein) for most zonation markers showed strong concordance between mRNA and protein. Although classical growth hormone regulated sex-biased gene products also exhibited concordance, ∼60% of sex-biased gene products showed protein-level enrichment without corresponding mRNA differences. In contrast, transition between feeding and starvation triggered widespread changes in mRNA expression without significantly affecting protein levels. In particular, key lipogenic mRNAs (e.g. Acly , Acaca , and Fasn ) were dramatically induced by feeding, but their corresponding proteins (ACLY, ACC1, and FAS) showed little to no change even as functional de novo lipogenic activity increased ∼28-fold in the fed state. To facilitate further exploration of these findings, we developed Discorda ( https://shinoda-lab.shinyapps.io/discorda/ ), a web database for interactive analysis. Our findings reinforce the principle that mRNA changes do not reliably predict corresponding protein levels (and vice versa), particularly in the context of sex and acute metabolic regulation of hepatocytes, and that de novo lipogenesis activity can be completely uncoupled from changes in protein expression.
Collapse
|
45
|
Mohammed Y, Richard VR, Reyes Madlangsakay MI, Lao Y, Spicer V, Popp R, Gaither C, Hennecken L, Kleinekofort W, Zahedi RP, Borchers CH. SysQuan: Repurposing SILAC Mice for the Cost-Effective Absolute Quantitation of the Human Proteome. Mol Cell Proteomics 2025; 24:100974. [PMID: 40254065 PMCID: PMC12143667 DOI: 10.1016/j.mcpro.2025.100974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/04/2025] [Accepted: 03/22/2025] [Indexed: 04/22/2025] Open
Abstract
Relative quantitation, used by most mass spectrometry-based proteomics laboratories to determine protein fold-changes, requires samples being processed and analyzed together for best comparability through minimizing batch differences. This limits the adoption of mass spectrometry-based proteomics in population-wide studies and the detection of subtle but relevant changes in heterogeneous samples. Absolute quantitation circumvents these limitations and enables comparison of results across laboratories, studies, and over time. However, high cost of the essential stable isotope labeled (SIL) standards prevents widespread access and limits the number of quantifiable proteins. Our new approach, called "SysQuan", repurposes SILAC mouse tissues/biofluids as system-wide internal standards for matched human samples to enable absolute quantitation of, theoretically, two-thirds of the human proteome using 157,086 shared tryptic peptides, of which 73,901 with lysine on the c terminus. We demonstrate that SysQuan enables quantification of 70% and 31% of the liver and plasma proteomes, respectively. We demonstrate for 14 metabolic proteins that abundant SIL mouse tissues enable cost-effective reverse absolute quantitation in, theoretically, 1000s of human samples. Moreover, 10,000s of light/heavy doublets in untargeted SysQuan datasets enable unique postacquisition absolute quantitation. SysQuan empowers researchers to replace relative quantitation with affordable absolute quantitation at scale, making data comparable across laboratories, diseases and tissues, enabling completely novel study designs and increasing reusability of data in repositories.
Collapse
Affiliation(s)
- Yassene Mohammed
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, Quebec, Canada; Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Vincent R Richard
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | | | - Ying Lao
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Robert Popp
- MRM Proteomics, Inc, Montreal, Quebec, Canada
| | - Claudia Gaither
- MRM Proteomics, Inc, Montreal, Quebec, Canada; Faculty of Veterinary Medicine, Department of Clinical Sciences, University of Montreal, Saint-Hyacinthe, Quebec, Canada
| | - Laura Hennecken
- Department of Engineering, Hochschule RheinMain, Rüsselsheim am Main, Germany
| | | | - René P Zahedi
- Manitoba Centre for Proteomics and Systems Biology, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada; Paul Abrechtsen Research Institute, CancerCare Manitoba, Winnipeg, Manitoba, Canada.
| | - Christoph H Borchers
- Segal Cancer Proteomics Centre, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, Quebec, Canada; Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada; Department of Pathology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
46
|
Gericke N, Beqaj D, Kronenberger T, Kulik A, Gavriilidou A, Franz-Wachtel M, Schoppmeier U, Harbig T, Rapp J, Grin I, Ziemert N, Link H, Nieselt K, Macek B, Wohlleben W, Stegmann E, Wagner S. Unveiling the substrate specificity of the ABC transporter Tba and its role in glycopeptide biosynthesis. iScience 2025; 28:112135. [PMID: 40171492 PMCID: PMC11960670 DOI: 10.1016/j.isci.2025.112135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/16/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
Glycopeptide antibiotics (GPA) such as vancomycin are essential last-resort antibiotics produced by actinomycetes. Their biosynthesis is encoded within biosynthetic gene clusters, also harboring genes for regulation, and transport. Diverse types of GPAs have been characterized that differ in peptide backbone composition and modification patterns. However, little is known about the ATP-binding cassette (ABC) transporters facilitating GPA export. Employing a multifaceted approach, we investigated the substrate specificity of GPA ABC-transporters toward the type-I GPA balhimycin. Phylogenetic analysis suggested and trans-complementation experiments confirmed that balhimycin is exported only by the related type I GPA transporters Tba and Tva (transporter of vancomycin). Molecular dynamics simulations and mutagenesis experiments showed that Tba exhibits specificity toward the peptide backbone rather than the modifications. Unexpectedly, deletion or functional inactivation of Tba halted balhimycin biosynthesis. Combined with proximity biotinylation experiments, this suggested that the interaction of the active transporter with the biosynthetic machinery is required for biosynthesis.
Collapse
Affiliation(s)
- Nicola Gericke
- Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Dardan Beqaj
- Microbial Active Compounds, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Thales Kronenberger
- Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- Partner-Site: DZIF Tübingen, Elfriede-Aulhorn-Str. 6/Auf der Morgenstelle 28, 72076 Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonrinne 3, 70211 Kuopio, Finland
| | - Andreas Kulik
- Microbial Active Compounds, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Athina Gavriilidou
- Translational Genome Mining for Natural Products, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Auf der Morgenstelle 24, 72076 Tübingen, Germany
| | - Mirita Franz-Wachtel
- Proteome Center Tübingen, Institute of Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Ulrich Schoppmeier
- Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), University of Tübingen, 72076 Tübingen, Germany
| | - Theresa Harbig
- Interfaculty Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Sand 14, 72076 Tübingen, Germany
| | - Johanna Rapp
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), University of Tübingen, 72076 Tübingen, Germany
- Bacterial Metabolomics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Iwan Grin
- Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Nadine Ziemert
- Partner-Site: DZIF Tübingen, Elfriede-Aulhorn-Str. 6/Auf der Morgenstelle 28, 72076 Tübingen, Germany
- Translational Genome Mining for Natural Products, Interfaculty Institute of Microbiology and Infection Medicine Tübingen (IMIT), University of Tübingen, Auf der Morgenstelle 24, 72076 Tübingen, Germany
| | - Hannes Link
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), University of Tübingen, 72076 Tübingen, Germany
- Bacterial Metabolomics, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Kay Nieselt
- Interfaculty Institute for Bioinformatics and Medical Informatics (IBMI), University of Tübingen, Sand 14, 72076 Tübingen, Germany
| | - Boris Macek
- Proteome Center Tübingen, Institute of Cell Biology, University of Tübingen, Auf der Morgenstelle 15, 72076 Tübingen, Germany
| | - Wolfgang Wohlleben
- Partner-Site: DZIF Tübingen, Elfriede-Aulhorn-Str. 6/Auf der Morgenstelle 28, 72076 Tübingen, Germany
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), University of Tübingen, 72076 Tübingen, Germany
- Microbiology/Biotechnology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
| | - Evi Stegmann
- Microbial Active Compounds, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Auf der Morgenstelle 28, 72076 Tübingen, Germany
- Partner-Site: DZIF Tübingen, Elfriede-Aulhorn-Str. 6/Auf der Morgenstelle 28, 72076 Tübingen, Germany
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), University of Tübingen, 72076 Tübingen, Germany
| | - Samuel Wagner
- Cellular and Molecular Microbiology, Interfaculty Institute of Microbiology and Infection Medicine (IMIT), University of Tübingen, Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
- Partner-Site: DZIF Tübingen, Elfriede-Aulhorn-Str. 6/Auf der Morgenstelle 28, 72076 Tübingen, Germany
- Excellence Cluster "Controlling Microbes to Fight Infections" (CMFI), University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
47
|
Li W, Dasgupta A, Yang K, Wang S, Hemandhar-Kumar N, Chepyala SR, Yarbro JM, Hu Z, Salovska B, Fornasiero EF, Peng J, Liu Y. Turnover atlas of proteome and phosphoproteome across mouse tissues and brain regions. Cell 2025; 188:2267-2287.e21. [PMID: 40118046 PMCID: PMC12033170 DOI: 10.1016/j.cell.2025.02.021] [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: 09/23/2024] [Revised: 01/27/2025] [Accepted: 02/21/2025] [Indexed: 03/23/2025]
Abstract
Understanding how proteins in different mammalian tissues are regulated is central to biology. Protein abundance, turnover, and post-translational modifications such as phosphorylation are key factors that determine tissue-specific proteome properties. However, these properties are challenging to study across tissues and remain poorly understood. Here, we present Turnover-PPT, a comprehensive resource mapping the abundance and lifetime of 11,000 proteins and 40,000 phosphosites in eight mouse tissues and various brain regions using advanced proteomics and stable isotope labeling. We reveal tissue-specific short- and long-lived proteins, strong correlations between interacting protein lifetimes, and distinct impacts of phosphorylation on protein turnover. Notably, we discover a remarkable pattern of turnover changes for peroxisome proteins in specific tissues and that phosphorylation regulates the stability of neurodegeneration-related proteins, such as Tau and α-synuclein. Thus, Turnover-PPT provides fundamental insights into protein stability, tissue dynamic proteotypes, and functional protein phosphorylation and is accessible via an interactive web-based portal at https://yslproteomics.shinyapps.io/tissuePPT.
Collapse
Affiliation(s)
- Wenxue Li
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | - Abhijit Dasgupta
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ka Yang
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shisheng Wang
- Department of General Surgery and Liver Transplant Center, Proteomics-Metabolomics Analysis Platform, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Nisha Hemandhar-Kumar
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Surendhar R Chepyala
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jay M Yarbro
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zhenyi Hu
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | - Barbora Salovska
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | - Eugenio F Fornasiero
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, 37073 Göttingen, Germany; Department of Life Sciences, University of Trieste, 34127 Trieste, Italy.
| | - Junmin Peng
- Departments of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA; Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA; Department of Biomedical Informatics & Data Science, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
48
|
Zhang C, Chen L. 6-Methoxyflavone inhibits glycolytic energy metabolism in HeLa cells. BMC Cancer 2025; 25:719. [PMID: 40247232 PMCID: PMC12004806 DOI: 10.1186/s12885-025-14133-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Enhanced glycolytic levels in cancer cells are a common characteristic of many cancer types. Modulation of glycolytic metabolism is crucial for enhancing the efficacy of cancer therapy. The specific role of 6-methoxyflavone in regulating glycolytic metabolism in cancer cells remains unclear. This study aimed to elucidate the impact of 6-methoxyflavone on glycolytic metabolism in cervical cancer cells and its clinical relevance. METHODS The tandem mass tag (TMT) proteomic analysis was used to identify significantly enriched biological processes and pathways in HeLa cells after treatment with 6-methoxyflavone. Additionally, the differential expression of glycolysis-related proteins was validated using parallel reaction monitoring (PRM) proteomics. Untargeted and targeted metabolomics analyses were used to identify differentially expressed glycolysis-related metabolites. Furthermore, alternative splicing, new transcripts, and domain analyses were used to detect the effects of 6-methoxyflavone on the structures of glycolysis-related genes and proteins. Subcellular localization, molecular docking, and non-covalent interaction analyses were used to detect the subcellular localization, affinity of 6-methoxyflavone for glycolysis-related proteins, and sites of non-covalent interactions. Clinical characteristics and immunological correlation analyses were used to elucidate the relationships between glycolysis-related genes and clinicopathological characteristics, survival, prognosis, and immune-related indicators of patients with cervical cancer. Finally, glycolysis stress tests and enzyme activity assays were used to verify the effect of 6-methoxyflavone on glycolysis in HeLa cells. RESULTS TMT and PRM proteomics, as well as untargeted and targeted metabolomics results, showed that 6-methoxyflavone downregulated the expression levels of glycolysis-related proteins and metabolites in HeLa cells, and that the structures and functions of glycolysis-related genes and proteins in the cytoplasm underwent changes. 6-Methoxyflavone had a good affinity for nine glycolysis-related proteins, all of which had non-covalent interaction sites. Clinical characteristics and immune correlation analyses showed relationships between 6-methoxyflavone and five clinical characteristics, survival prognosis, and four immune-related indicators in patients with cervical cancer. After treatment with 6-methoxyflavone, the basal glycolytic level, maximum glycolytic capacity, and glycolytic reserve of HeLa cells were downregulated. Additionally, 6-methoxyflavone inhibited the activity of pyruvate kinase. CONCLUSION 6-Methoxyflavone inhibited energy metabolism in HeLa cells through the glycolysis pathway. 6-Methoxyflavone may be related to five clinical characteristics, prognosis, tumor microenvironment, immune cells, immune checkpoints, and immunotherapy efficacy in patients with cervical cancer.
Collapse
Affiliation(s)
- Chaihong Zhang
- Department of Obstetrics and Gynecology, Shaanxi Provincial People's Hospital, 256 Youyi West Road, Xi'an City, Shaanxi Province, 710000, China
| | - Lihong Chen
- Department of Obstetrics and Gynecology, Shaanxi Provincial People's Hospital, 256 Youyi West Road, Xi'an City, Shaanxi Province, 710000, China.
| |
Collapse
|
49
|
van der Hoorn D, Lauria F, Chaytow H, Faller KME, Huang YT, Kline RA, Signoria I, Morris K, Wishart TM, Groen EJN, Viero G, Gillingwater TH. Dynamic modulation of the motor neuron translatome during developmental synapse elimination. Sci Signal 2025; 18:eadr0176. [PMID: 40233176 DOI: 10.1126/scisignal.adr0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/25/2025] [Indexed: 04/17/2025]
Abstract
The developmental sculpting of neuromuscular circuitry in early postnatal life occurs through the process of synapse elimination: Supernumerary axon inputs are gradually eliminated from the neuromuscular junction (NMJ), resulting in each muscle fiber being innervated by a single axon terminal. Here, we investigated the molecular pathways underlying this process using a ChAT-RiboTag mouse model in which we isolated ribosome-bound mRNAs in motor neurons during synapse elimination in vivo. Analysis of these mRNAs using translating ribosome affinity purification followed by RNA sequencing (TRAP-seq) revealed dynamic changes in the motor neuron translatome over the first 2 weeks of life, which were largely independent of parallel transcriptional changes and correlated with the progressive elimination of supernumerary inputs. Bioinformatic analysis identified distinct clusters of transcripts that were translated at specific time points during synapse elimination. Treating mice with two small molecules that were predicted to independently target the proteins or pathways encoded by the transcript cluster associated with neural metabolism increased the rate of synapse elimination in vivo. Together, these data provide a cell type-specific overview of temporal modifications occurring in the motor neuron translatome during synapse elimination, revealing rapid and dynamic responses to postnatal developmental cues.
Collapse
Affiliation(s)
- Dinja van der Hoorn
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Fabio Lauria
- Institute of Biophysics, CNR Unit at Trento, Trento, Italy
| | - Helena Chaytow
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Kiterie M E Faller
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Yu-Ting Huang
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Rachel A Kline
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Ilaria Signoria
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, Netherlands
| | - Kim Morris
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Thomas M Wishart
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
- Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Ewout J N Groen
- UMC Utrecht Brain Center, Department of Neurology and Neurosurgery, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Thomas H Gillingwater
- Edinburgh Medical School: Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
50
|
Paternoga H, Xia L, Dimitrova-Paternoga L, Li S, Yan LL, Oestereich M, Kasvandik S, Nanjaraj Urs AN, Beckert B, Tenson T, Zaher H, Inada T, Wilson DN. Structure of a Gcn2 dimer in complex with the large 60S ribosomal subunit. Proc Natl Acad Sci U S A 2025; 122:e2415807122. [PMID: 40198700 PMCID: PMC12012509 DOI: 10.1073/pnas.2415807122] [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: 08/05/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
The integrated stress response (ISR) is a central signaling network that enables eukaryotic cells to respond to a variety of different environmental stresses. Such stresses cause ribosome collisions that lead to activation of the kinase Gcn2, resulting in the phosphorylation and inactivation of eukaryotic initiation factor 2 and thereby promoting selective translation of mRNAs to restore homeostasis. Despite the importance of the ISR and intensive study over the past decades, structural insight into how Gcn2 interacts with ribosomal particles has been lacking. Using ex vivo affinity purification approaches, we have obtained a cryoelectron microscopy structure of a yeast Gcn2 dimer in complex with the ribosomal 60S subunit. The Gcn2 dimer is formed by dimerization of the histidine tRNA synthetase-like domains, which establish extensive interactions with the stalk-base and sarcin-ricin loop of the 60S subunit. The C-terminal domain of Gcn2 is also dimerized and occupies the A- and P-site tRNA binding sites at the peptidyl-transferase center of the 60S subunit. Complementary functional studies indicate that binding of Gcn2 to the 60S subunit does not require the coactivators Gcn1 or Gcn20, nor does it lead to phosphorylation of eIF2α. Instead, upon stress, we observe a shift of Gcn2 from the 60S subunit into the colliding ribosome fraction, suggesting that the Gcn2-60S complex represents an inactive stand-by state to enable a rapid redistribution to collided ribosomes, and thereby facilitating a quick and efficient response to stress.
Collapse
Affiliation(s)
- Helge Paternoga
- Department of Chemistry, Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg20146, Germany
| | - Lu Xia
- Division of Ribonucleic Acid (RNA) and Gene Regulation, Institute of Medical Science, The University of Tokyo, Minato-Ku, Tokyo108-8639, Japan
| | - Lyudmila Dimitrova-Paternoga
- Department of Chemistry, Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg20146, Germany
| | - Sihan Li
- Division of Ribonucleic Acid (RNA) and Gene Regulation, Institute of Medical Science, The University of Tokyo, Minato-Ku, Tokyo108-8639, Japan
| | - Liewei L. Yan
- Department of Biology, Washington University in St. Louis, St. Louis, MO63130
| | - Malte Oestereich
- Department of Chemistry, Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg20146, Germany
| | - Sergo Kasvandik
- Faculty of Science and Technology, Institute of Technology, University of Tartu, Tartu50411, Estonia
| | | | - Bertrand Beckert
- Dubochet Center for Imaging at the Ecole Polytechnique Fédérale de Lausanne and the Université de Lausanne (DCI EPFL-UNIL), Quartier UNIL-Sorge, Bâtiment Génopode, Lausanne1015, Switzerland
| | - Tanel Tenson
- Faculty of Science and Technology, Institute of Technology, University of Tartu, Tartu50411, Estonia
| | - Hani Zaher
- Department of Biology, Washington University in St. Louis, St. Louis, MO63130
| | - Toshifumi Inada
- Division of Ribonucleic Acid (RNA) and Gene Regulation, Institute of Medical Science, The University of Tokyo, Minato-Ku, Tokyo108-8639, Japan
| | - Daniel N. Wilson
- Department of Chemistry, Institute for Biochemistry and Molecular Biology, University of Hamburg, Hamburg20146, Germany
| |
Collapse
|