101
|
Daliri K, Hescheler J, Newby GA, Clement K, Liu DR, Pfannkuche K. Modulating Collagen I Expression in Fibroblasts by CRISPR-Cas9 Base Editing of the Collagen 1A1 Promoter. Int J Mol Sci 2025; 26:3041. [PMID: 40243657 PMCID: PMC11989027 DOI: 10.3390/ijms26073041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Fibrotic diseases, contributing to a significant portion of global mortality, highlight the need for innovative therapies. This study explores a novel approach to disrupt the expression of collagen by using adenine base editing to target Col1a1, a key gene driving both fibrosis and cancer metastasis. Editing Col1a1 in fibroblasts demonstrated 18% editing efficiency. An analysis of a specific clone harboring a CCAAT-to-CCGGA mutation in the Col1a1 promoter revealed reduced collagen production. Notably, when wild-type fibroblasts were cultured on the Col1a1-edited matrix, no compensatory collagen upregulation was detected, suggesting a lack of feedback mechanism in fibroblasts. Furthermore, the matrix derived from edited fibroblasts did not support the growth of MCF-7 cancer cells. These findings suggest that Col1a1 gene editing holds promise as a potential therapeutic strategy for fibrotic diseases. Further investigation is warranted to fully elucidate the implications of these findings for fibrosis and cancer.
Collapse
Affiliation(s)
- Karim Daliri
- Centre for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty and University Hospital of Cologne, 50931 Cologne, Germany
- Marga and Walter Boll-Laboratory for Cardiac Tissue Engineering, University of Cologne, 50931 Cologne, Germany
| | - Jürgen Hescheler
- Centre for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty and University Hospital of Cologne, 50931 Cologne, Germany
| | - Gregory A. Newby
- McKusick-Nathans Institute and Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kendell Clement
- Department of Biomedical Informatics, University of Utah, Salt Lake City, UT 84108, USA;
| | - David R. Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA;
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA
| | - Kurt Pfannkuche
- Centre for Physiology and Pathophysiology, Institute for Neurophysiology, University of Cologne, Medical Faculty and University Hospital of Cologne, 50931 Cologne, Germany
- Marga and Walter Boll-Laboratory for Cardiac Tissue Engineering, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
102
|
Meert L, Pelicano de Almeida M, Dekker MR, Dekkers DHW, Nowosad K, Huylebroeck D, van den Hout M, Ozgür Z, van IJcken WFJ, Demmers J, Fornerod M, Poot RA. A CHD8-TRRAP axis facilitates MYC and E2F target gene regulation in human neural stem cells. iScience 2025; 28:111978. [PMID: 40104050 PMCID: PMC11914185 DOI: 10.1016/j.isci.2025.111978] [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: 11/07/2023] [Revised: 11/06/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025] Open
Abstract
Mutations in ATP-dependent chromatin remodeler CHD8 cause one of the most frequent monogenetic forms of autism and are associated with brain overgrowth. Nevertheless, the activities of CHD8 in autism-relevant cell types are still poorly understood. Here, we purify the CHD8 protein from human neural stem cells and determine its interaction partners using mass spectrometry. We identify the TRRAP complex, a coactivator of MYC and E2F transcription factors, as a prominent CHD8 interaction partner. CHD8 colocalizes genome-wide with TRRAP and binds together at MYC and E2F target gene promoters in human neural stem cells. Depletion of CHD8 or TRRAP in human neural stem cells shows downregulation of MYC and E2F target genes as the most prominent gene-regulatory events. Depletion of CHD8 diminishes cell-cycle entry into S-phase. MYC and E2F transcription factors are established oncogenes and regulate cell growth. Our results link CHD8 to TRRAP in facilitating the regulation of MYC and E2F target genes in human neural stem cells.
Collapse
Affiliation(s)
- Lize Meert
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | | | - Mike R Dekker
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Dick H W Dekkers
- Center for Proteomics, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Karol Nowosad
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | | | - Zeliha Ozgür
- Center for Biomics, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
- Center for Biomics, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Jeroen Demmers
- Center for Proteomics, Erasmus MC, 3015 CN Rotterdam, the Netherlands
| | - Maarten Fornerod
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| | - Raymond A Poot
- Department of Cell Biology, Erasmus MC, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands
| |
Collapse
|
103
|
Fahrion J, Gupta S, Mastroleo F, Dussap CG, Leys N. Chronic low-dose rate irradiation induces transient hormesis effect on cyanobacterium Limnospira indica. iScience 2025; 28:111891. [PMID: 40124510 PMCID: PMC11926712 DOI: 10.1016/j.isci.2025.111891] [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: 07/12/2024] [Revised: 08/27/2024] [Accepted: 01/22/2025] [Indexed: 03/25/2025] Open
Abstract
Cultures of Limnospira indica were exposed to low-dose rate γ-irradiation for 8 weeks to simulate 2 months of a Mars transit irradiation. Two experiments were conducted: in the first, 5% v/v inoculations were used over 2-week batches; in the second, 25% v/v inoculations over 1-week batches. The cultures were continuously illuminated (45 μmol photons m-2 s-1, LEDs). A transient hormesis effect was observed in experiment 1, with irradiated cultures showing higher dry weight (1.88 ± 0.05 g L-1) than controls (1.70 ± 0.06 g L-1) on day 14. Irradiated cultures also had fewer pigments. Experiment 2 showed similar, though less pronounced, results. These findings suggest that Limnospira indica would not be negatively affected by cosmic radiation during Mars transit, though further validation under space flight conditions is needed. The resilience of Limnospira indica to chronic low-dose radiation supports its potential for oxygen and food production in life support systems for manned space missions.
Collapse
Affiliation(s)
- Jana Fahrion
- Microbial Biotechnology Unit, Nuclear Medical Applications, Belgian Nuclear Research Center SCK CEN, 2400 Mol, Belgium
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal, 63100 Clermont-Ferrand, France
| | - Surya Gupta
- Microbial Biotechnology Unit, Nuclear Medical Applications, Belgian Nuclear Research Center SCK CEN, 2400 Mol, Belgium
| | - Felice Mastroleo
- Microbial Biotechnology Unit, Nuclear Medical Applications, Belgian Nuclear Research Center SCK CEN, 2400 Mol, Belgium
| | - Claude Gilles Dussap
- Université Clermont Auvergne, CNRS, Clermont Auvergne INP, Institut Pascal, 63100 Clermont-Ferrand, France
| | - Natalie Leys
- Microbial Biotechnology Unit, Nuclear Medical Applications, Belgian Nuclear Research Center SCK CEN, 2400 Mol, Belgium
| |
Collapse
|
104
|
Decker M, Huang BJ, Ware T, Boone C, Tang M, Ybarra J, Ballapuram AC, Taran KA, Chen PY, Amendáriz M, Leung CJ, Harris M, Tjoa K, Hongo H, Abelson S, Rivera J, Ngo N, Herbst DM, Suciu RM, Guijas C, Sedighi K, Andalis T, Roche E, Xie B, Liu Y, Smith CC, Stieglitz E, Niphakis MJ, Cravatt BF, Shannon K. In vivo validation of the palmitoylation cycle as a therapeutic target in NRAS-mutant cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644389. [PMID: 40166265 PMCID: PMC11957127 DOI: 10.1101/2025.03.20.644389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Normal and oncogenic Ras proteins are functionally dependent on one or more lipid modifications1,2. Whereas K-Ras4b farnesylation is sufficient for stable association with the plasma membrane, farnesylated H-Ras, K-Ras4a, and N-Ras traffic to the Golgi where they must undergo palmitoylation before regulated translocation to cell membranes. N-Ras palmitoylation by the DHHC family of palmitoyl acyl transferases (PATs) and depalmitoylation by ABHD17 serine hydrolases is a dynamic process that is essential for the growth of acute myeloid leukemias (AMLs) harboring oncogenic NRAS mutations3-6. Here, we have tested whether co-targeting ABHD17 enzymes and Ras signal output would cooperatively inhibit the proliferation and survival of NRAS-mutant AMLs while sparing normal tissues that retain K-Ras4b function. We show that ABD778, a potent and selective ABHD17 inhibitor with in vivo activity, selectively reduces the growth of NRAS-mutant AML cells in vitro and is synergistic with the allosteric MEK inhibitor PD0325901 (PD901)7,8. Similarly, ABD778 and PD901 significantly extended the survival of recipient mice transplanted with three independent primary mouse AMLs harboring an oncogenic Nras G12D driver mutation. Resistant leukemias that emerged during continuous drug treatment acquired by-pass mutations that confer adaptive drug resistance and increase mitogen activated protein kinase (MAPK) signal output. ABD778 augmented the anti-leukemia activity of the pan-PI3 kinase inhibitor pictilisib9, the K/N-RasG12C inhibitor sotorasib10, and the FLT3 inhibitor gilteritinib11. Co-treatment with ABD778 and gilteritinib restored drug sensitivity in a patient-derived xenograft model of adaptive resistance to FLT3 inhibition. These data validate the palmitoylation cycle as a promising therapeutic target in AML and support exploring it in other NRAS-mutant cancers.
Collapse
Affiliation(s)
- Matthew Decker
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Benjamin J. Huang
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Timothy Ware
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher Boone
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Michelle Tang
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Julia Ybarra
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Katrine A. Taran
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Pan-Yu Chen
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Marcos Amendáriz
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Camille J. Leung
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Max Harris
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Karensa Tjoa
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Henry Hongo
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Sydney Abelson
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jose Rivera
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Nhi Ngo
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | | | - Radu M. Suciu
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Carlos Guijas
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Kimia Sedighi
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Taylor Andalis
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Elysia Roche
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Boer Xie
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Yunlong Liu
- Center for Center for Medical Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Catherine C. Smith
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Elliot Stieglitz
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Kevin Shannon
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
105
|
Li Y, Wang J, Zhou L, Gu W, Qin L, Peng D, Li S, Zheng D, Wu Q, Long Y, Yao Y, Lin S, Sun M, Zhang X, Wang J, Liu P, Kong X, Li P. DNMT1 inhibition reprograms T cells to NK-like cells with potent antitumor activity. Sci Immunol 2025; 10:eadm8251. [PMID: 40117344 DOI: 10.1126/sciimmunol.adm8251] [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: 11/07/2023] [Revised: 11/20/2024] [Accepted: 02/13/2025] [Indexed: 03/23/2025]
Abstract
Inactivation of the transcription factor BCL11B reprograms T cells into induced-T-to-NK cells (ITNKs). However, it remains unclear how BCL11B suppresses natural killer (NK) cell transcriptional programs. Here, we identified that the DNA methyltransferase DNMT1 physically interacts with BCL11B, increasing BCL11B stability and the fidelity of DNA methylation maintenance for NK cell-related genes, thereby repressing their expression. Moreover, DNMT1 maintains the epigenetic silencing of a distinct subset of NK cell-related genes independent of BCL11B. DNMT1 inhibition or depletion reprograms T cells and chimeric antigen receptor (CAR)-T cells into NK-like cells that exhibit more robust antitumor effects than BCL11B-deficient ITNKs and parental CAR-T cells. Moreover, H3K27me3 (trimethylation of histone 3 lysine 27) synergizes with DNA methylation to repress NK cell-related pathways, and combined EZH2 (enhancer of zeste homolog 2) and DNMT1 inhibition potentiates both the reprogramming and cytotoxicity of NK-like cells. Our findings uncover the molecular mechanisms that safeguard T cell identity and provide a rationale for deriving NK-like cells with epigenetic inhibitors for cancer immunotherapy.
Collapse
Affiliation(s)
- Yao Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jiongliang Wang
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Linfu Zhou
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Wenbin Gu
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Le Qin
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Guangdong Zhaotai Cell Bioscience Ltd., Shunde, China
| | - Dongdong Peng
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shanglin Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Diwei Zheng
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Qiting Wu
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Youguo Long
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Yao Yao
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Shouheng Lin
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Mingwei Sun
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, China
| | - Xiaofei Zhang
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jie Wang
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Pentao Liu
- School of Biomedical Sciences, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong SAR, China
| | - Xiangqian Kong
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Peng Li
- China-New Zealand Joint Laboratory on Biomedicine and Health, National Key Laboratory of Immune Response and Immunotherapy, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China
- Department of Surgery, Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
106
|
Hermanns T, Kolek S, Uthoff M, de Heiden RA, Mulder MPC, Baumann U, Hofmann K. A family of bacterial Josephin-like deubiquitinases with an irreversible cleavage mode. Mol Cell 2025; 85:1202-1215.e5. [PMID: 40037356 DOI: 10.1016/j.molcel.2025.02.002] [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: 07/24/2024] [Revised: 12/05/2024] [Accepted: 02/04/2025] [Indexed: 03/06/2025]
Abstract
Many intracellular bacteria secrete deubiquitinase (DUB) effectors into eukaryotic host cells to keep the bacterial surface or the enclosing vesicle membrane free of ubiquitin marks. This study describes a family of DUBs from several bacterial genera, including Simkania, Parachlamydia, Burkholderia, and Pigmentiphaga, which is structurally related to eukaryotic Josephin-type DUBs but contains members that catalyze a unique destructive substrate deubiquitination. These ubiquitin C-terminal clippases (UCCs) cleave ubiquitin before the C-terminal diGly motif, thereby truncating the modifier and leaving a remnant on the substrate. By comparing the crystal structures of substrate-bound clippases and a closely related conventional DUB, we identified the factors causing this shift and found them to be conserved in other clippases, including one highly specific for M1-linked ubiquitin chains. This enzyme class has great potential to serve as tools for studying the ubiquitin system, particularly aspects involving branched chains.
Collapse
Affiliation(s)
- Thomas Hermanns
- Institute for Genetics, University of Cologne, Zülpicher Straße 47a, 50674 Cologne, Germany
| | - Susanne Kolek
- Institute for Genetics, University of Cologne, Zülpicher Straße 47a, 50674 Cologne, Germany
| | - Matthias Uthoff
- Institute of Biochemistry, University of Cologne, Zülpicher Straße 47, 50674 Cologne, Germany
| | - Richard A de Heiden
- Department of Cell and Chemical Biology, Leiden University Medical Center (LUMC), Einthovenweg 20, 2333ZC Leiden, the Netherlands
| | - Monique P C Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center (LUMC), Einthovenweg 20, 2333ZC Leiden, the Netherlands
| | - Ulrich Baumann
- Institute of Biochemistry, University of Cologne, Zülpicher Straße 47, 50674 Cologne, Germany
| | - Kay Hofmann
- Institute for Genetics, University of Cologne, Zülpicher Straße 47a, 50674 Cologne, Germany.
| |
Collapse
|
107
|
Wang J, Zhang Y, Wang J, Wang L, Li B, Liu S, Sun X, Ma Z. Fungal chromatin remodeler Isw1 modulates translation via regulating tRNA transcription. Nucleic Acids Res 2025; 53:gkaf225. [PMID: 40167328 PMCID: PMC11959538 DOI: 10.1093/nar/gkaf225] [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: 09/17/2024] [Revised: 02/15/2025] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
Chromatin dynamics are essential for regulating DNA processes in response to environmental stimuli. Although ISWI-family enzymes are known to remodel chromatin by sliding nucleosomes in budding yeast, their functional roles and outputs in eukaryotes remain largely unknown. In this study, we investigated chromatin accessibility in the phytopathogenic fungus Fusarium graminearum treated with and without putrescine, a compound that rapidly induces the biosynthesis of the mycotoxin deoxynivalenol (DON). Putrescine globally alters chromatin accessibility, with the ATP-dependent chromatin remodeler FgIsw1 emerging as a key regulator. Unexpectedly, deletion of FgIsw1 did not affect the transcription of DON biosynthesis genes (Tri) but significantly disrupted transfer RNA (tRNA) transcription, leading to a dramatic decline in translation of DON biosynthesis enzymes. Mechanistically, FgIsw1 maintains nucleosome phasing in tRNA chromatin regions, ensuring efficient tRNA transcription. As a result, ΔFgIsw1 was unable to produce DON and lost its virulence on the host plant. These results highlight a novel role of chromatin remodelers in regulating protein translation through the control of tRNA transcription.
Collapse
Affiliation(s)
- Jing Wang
- Department of Plant Pathology, College of Plant Protection, Southwest University, Chongqing 400715, China
- National Key Laboratory of Rice Biological Breeding, Institute of Biotechnology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Yueqi Zhang
- National Key Laboratory of Rice Biological Breeding, Institute of Biotechnology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jingrui Wang
- National Key Laboratory of Rice Biological Breeding, Institute of Biotechnology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Liuqin Wang
- National Key Laboratory of Rice Biological Breeding, Institute of Biotechnology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Binshan Li
- Department of Plant Pathology, College of Plant Protection, Southwest University, Chongqing 400715, China
| | - Shuang Liu
- Department of Plant Pathology, College of Plant Protection, Southwest University, Chongqing 400715, China
| | - Xuepeng Sun
- Collaborative Innovation Center for Efficient and Green Production of Agriculture in Mountainous Areas of Zhejiang Province, College of Horticulture Science, Zhejiang A&F University, Hangzhou 311300, China
| | - Zhonghua Ma
- National Key Laboratory of Rice Biological Breeding, Institute of Biotechnology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
- Yazhouwan National Laboratory, Sanya 572025, China
| |
Collapse
|
108
|
Kavaklioglu G, Podhornik A, Vcelkova T, Marjanovic J, Beck MA, Phan-Canh T, Mair T, Miccolo C, Drino A, Doni M, Egger G, Chiocca S, Modic M, Seiser C. The domesticated transposon protein L1TD1 associates with its ancestor L1 ORF1p to promote LINE-1 retrotransposition. eLife 2025; 13:RP96850. [PMID: 40112032 PMCID: PMC11925450 DOI: 10.7554/elife.96850] [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] [Indexed: 03/22/2025] Open
Abstract
Repression of retrotransposition is crucial for the successful fitness of a mammalian organism. The domesticated transposon protein L1TD1, derived from LINE-1 (L1) ORF1p, is an RNA-binding protein that is expressed only in some cancers and early embryogenesis. In human embryonic stem cells, it is found to be essential for maintaining pluripotency. In cancer, L1TD1 expression is highly correlative with malignancy progression and as such considered a potential prognostic factor for tumors. However, its molecular role in cancer remains largely unknown. Our findings reveal that DNA hypomethylation induces the expression of L1TD1 in HAP1 human tumor cells. L1TD1 depletion significantly modulates both the proteome and transcriptome and thereby reduces cell viability. Notably, L1TD1 associates with L1 transcripts and interacts with L1 ORF1p protein, thereby facilitating L1 retrotransposition. Our data suggest that L1TD1 collaborates with its ancestral L1 ORF1p as an RNA chaperone, ensuring the efficient retrotransposition of L1 retrotransposons, rather than directly impacting the abundance of L1TD1 targets. In this way, L1TD1 might have an important role not only during early development but also in tumorigenesis.
Collapse
Affiliation(s)
- Gülnihal Kavaklioglu
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of ViennaViennaAustria
| | - Alexandra Podhornik
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of ViennaViennaAustria
| | - Terezia Vcelkova
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of ViennaViennaAustria
| | - Jelena Marjanovic
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of ViennaViennaAustria
| | - Mirjam A Beck
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of ViennaViennaAustria
| | - Trinh Phan-Canh
- Medical University of Vienna, Max Perutz Labs Vienna, Campus Vienna BiocenterViennaAustria
| | - Theresia Mair
- Department of Pathology, Medical University of ViennaViennaAustria
| | - Claudia Miccolo
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCSMilanItaly
| | - Aleksej Drino
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of ViennaViennaAustria
| | - Mirko Doni
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCSMilanItaly
| | - Gerda Egger
- Department of Pathology, Medical University of ViennaViennaAustria
| | - Susanna Chiocca
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCSMilanItaly
| | - Miha Modic
- National Institute of ChemistryLjubljanaSlovenia
- Dementia Research Institute at King’s College London and The Francis Crick instituteLondonUnited Kingdom
| | - Christian Seiser
- Division of Cell and Developmental Biology, Center for Anatomy and Cell Biology, Medical University of ViennaViennaAustria
| |
Collapse
|
109
|
Manavski N, Schwenkert S, Kunz HH, Leister D, Meurer J. Targeted translation inhibition of chloroplast and mitochondrial mRNAs by designer pentatricopeptide repeat proteins. Nucleic Acids Res 2025; 53:gkaf222. [PMID: 40138717 PMCID: PMC11941472 DOI: 10.1093/nar/gkaf222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/03/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Pentatricopeptide repeat (PPR) proteins are crucial for organellar gene expression. To establish a tool for gene expression manipulation in Arabidopsis plastids and genetically inaccessible mitochondria, we engineered designer (dPPR) proteins to specifically inhibit the translation of organellar mRNAs by masking their start codons. Unlike prior methods for targeted downregulation of gene expression, which rely on re-targeting native PPR proteins to RNA sequences closely related to their original targets, our approach employs a synthetic P-type PPR scaffold that can be designed to bind any RNA sequence of interest. Here, using dPPR-psbK and dPPR-nad7, we targeted the psbK mRNA in chloroplasts and the nad7 mRNA in mitochondria, respectively. dPPR-psbK effectively bound to psbK mRNA and inhibited its translation with high specificity, resulting in disrupted PSII supercomplexes and reduced photosynthetic efficiency. dPPR-nad7 suppressed nad7 translation, affecting NADH oxidase activity in complex I and growth retardation. Comparing phenotypes with tobacco psbK knockouts and nad7 knockdown bir6-2 mutants, along with quantitative proteomics, showed no clear evidence of physiologically relevant off-target effects. Our findings establish dPPR proteins as precise tools for targeted translation inhibition, facilitating functional studies of organellar genes and offering a novel approach with potential for manipulating organellar gene expression in diverse plant species.
Collapse
Affiliation(s)
- Nikolay Manavski
- Plant Molecular Biology, Faculty of Biology, Ludwig-Maximilians-Universität Munich, Großhaderner Street 2-4, 82152 Planegg-Martinsried, Germany
| | - Serena Schwenkert
- Plant Molecular Biology, Faculty of Biology, Ludwig-Maximilians-Universität Munich, Großhaderner Street 2-4, 82152 Planegg-Martinsried, Germany
| | - Hans-Henning Kunz
- Plant Biochemistry, Faculty of Biology, Ludwig-Maximilians-Universität Munich, Großhaderner Street 2-4, 82152 Planegg-Martinsried, Germany
| | - Dario Leister
- Plant Molecular Biology, Faculty of Biology, Ludwig-Maximilians-Universität Munich, Großhaderner Street 2-4, 82152 Planegg-Martinsried, Germany
| | - Jörg Meurer
- Plant Molecular Biology, Faculty of Biology, Ludwig-Maximilians-Universität Munich, Großhaderner Street 2-4, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
110
|
Aval SF, Seffouh A, Moon KM, Foster LJ, Ortega J, Fredrick K. Role of the sarcin-ricin loop of 23S rRNA in biogenesis of the 50S ribosomal subunit. RNA (NEW YORK, N.Y.) 2025; 31:585-599. [PMID: 39875174 PMCID: PMC11912913 DOI: 10.1261/rna.080335.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/11/2025] [Indexed: 01/30/2025]
Abstract
The sarcin-ricin loop (SRL) is one of the most conserved segments of ribosomal RNA (rRNA). Translational GTPases (trGTPases), such as EF-G, EF-Tu, and IF2, form contacts with the SRL that are critical for GTP hydrolysis and factor function. Previous studies showed that expression of 23S rRNA lacking the SRL confers a dominant lethal phenotype in Escherichia coli Isolated ΔSRL particles were found to be not only inactive in protein synthesis but also incompletely assembled. In particular, block 4 of the subunit, which includes the peptidyl transferase center, remained unfolded. Here, we explore the basis of this assembly defect. We find that 23S rRNA extracted from ΔSRL subunits can be efficiently reconstituted into 50S subunits, and these reconstituted ΔSRL particles exhibit full peptidyl transferase activity. We also further characterize ΔSRL particles purified from cells, using cryo-EM and proteomic methods. These particles lack density for rRNA and r-proteins of block 4, consistent with earlier chemical probing data. Incubation of these particles with excess total r-protein of the large subunit (TP50) fails to restore substantial peptidyl transferase activity. Interestingly, proteomic analysis of control and mutant particles shows an overrepresentation of multiple assembly factors in the ΔSRL case. We propose that one or more GTPases normally act to release assembly factors, and this activity is blocked in the absence of the SRL.
Collapse
MESH Headings
- RNA, Ribosomal, 23S/chemistry
- RNA, Ribosomal, 23S/genetics
- RNA, Ribosomal, 23S/metabolism
- Escherichia coli/genetics
- Escherichia coli/metabolism
- Ribosome Subunits, Large, Bacterial/metabolism
- Ribosome Subunits, Large, Bacterial/genetics
- Escherichia coli Proteins/metabolism
- Escherichia coli Proteins/genetics
- Peptidyl Transferases/metabolism
- Peptidyl Transferases/genetics
- Cryoelectron Microscopy
- Nucleic Acid Conformation
- Protein Biosynthesis
- GTP Phosphohydrolases/metabolism
Collapse
Affiliation(s)
- Sepideh Fakhretaha Aval
- Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio 43210, USA
- Center for RNA Biology, The Ohio State University, Columbus, Ohio 43210, USA
| | - Amal Seffouh
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
- Centre for Structural Biology, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - Kyung-Mee Moon
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T1Z4, Canada
| | - Leonard J Foster
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T1Z4, Canada
| | - Joaquin Ortega
- Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
- Centre for Structural Biology, McGill University, Montreal, Quebec H3G 0B1, Canada
| | - Kurt Fredrick
- Ohio State Biochemistry Program, The Ohio State University, Columbus, Ohio 43210, USA
- Center for RNA Biology, The Ohio State University, Columbus, Ohio 43210, USA
- Department of Microbiology, The Ohio State University, Columbus, Ohio 43210, USA
| |
Collapse
|
111
|
Korek M, Mehta D, Uhrig GR, Daszkowska-Golec A, Novak O, Buchcik W, Marzec M. Strigolactone insensitivity affects the hormonal homeostasis in barley. Sci Rep 2025; 15:9375. [PMID: 40102576 PMCID: PMC11920428 DOI: 10.1038/s41598-025-94430-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 03/13/2025] [Indexed: 03/20/2025] Open
Abstract
In response to environmental changes, plants continuously make architectural changes in order to optimize their growth and development. The regulation of plant branching, influenced by environmental conditions and affecting hormone balance and gene expression, is crucial for agronomic purposes due to its direct correlation with yield. Strigolactones (SL), the youngest class of phytohormones, function to shape the architecture of plants by inhibiting axillary outgrowth. Barley plants harboring the mutation in the HvDWARF14 (HvD14) gene, which encodes the SL-specific receptor, produce almost twice as many tillers as wild-type (WT) Sebastian plants. Here, through hormone profiling and comparison of transcriptomic and proteomic changes between 2- and 4-week-old plants of WT and hvd14 genotypes, we elucidate a regulatory mechanism that might affect the tillering of SL-insensitive plants. The analysis showed statistically significant increased cytokinin content and decreased auxin and abscisic acid content in 'bushy' hvd14 compared to WT, which aligns with the commonly known actions of these hormones regarding branching regulation. Further, transcriptomic and proteomic analysis revealed a set of differentially expressed genes (DEG) and abundant proteins (DAP), among which 11.6% and 14.6% were associated with phytohormone-related processes, respectively. Bioinformatics analyses then identified a series of potential SL-dependent transcription factors (TF), which may control the differences observed in the hvd14 transcriptome and proteome. Comparison to available Arabidopsis thaliana data implicates a sub-selection of these TF as being involved in the transduction of SL signal in both monocotyledonous and dicotyledonous plants.
Collapse
Affiliation(s)
- Magdalena Korek
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Jagiellonska 28, 40-032, Katowice, Poland.
| | - Devang Mehta
- Department of Biological Sciences, University of Alberta, 11455 Saskatchewan Drive, Edmonton, AB, T6G 2E9, Canada
| | - Glen R Uhrig
- Department of Biological Sciences, University of Alberta, 11455 Saskatchewan Drive, Edmonton, AB, T6G 2E9, Canada
| | - Agata Daszkowska-Golec
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Jagiellonska 28, 40-032, Katowice, Poland
| | - Ondrej Novak
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany, The Czech Academy of Sciences, Olomouc, Czech Republic
| | - Weronika Buchcik
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Jagiellonska 28, 40-032, Katowice, Poland
| | - Marek Marzec
- Institute of Biology, Biotechnology and Environmental Protection, Faculty of Natural Sciences, University of Silesia in Katowice, Jagiellonska 28, 40-032, Katowice, Poland
| |
Collapse
|
112
|
Zhu J, Xie R, Ren Q, Zhou J, Chen C, Xie MX, Zhou Y, Zhang Y, Liu N, Wang J, Zhang Z, Liu X, Yan W, Gong Q, Dong L, Zhu J, Wang F, Xie Z. Asgard Arf GTPases can act as membrane-associating molecular switches with the potential to function in organelle biogenesis. Nat Commun 2025; 16:2622. [PMID: 40097441 PMCID: PMC11914678 DOI: 10.1038/s41467-025-57902-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 03/06/2025] [Indexed: 03/19/2025] Open
Abstract
Inward membrane budding, i.e., the bending of membrane towards the cytosol, is essential for forming and maintaining eukaryotic organelles. In eukaryotes, Arf GTPases initiate this inward budding. Our research shows that Asgard archaea genomes encode putative Arf proteins (AArfs). AArfs possess structural elements characteristic of their eukaryotic counterparts. When expressed in yeast and mammalian cells, some AArfs displayed GTP-dependent membrane targeting. In vitro, AArf associated with both eukaryotic and archaeal membranes. In yeast, AArfs interacted with and were regulated by key organelle biogenesis players. Expressing an AArf led to a massive proliferation of endomembrane organelles including the endoplasmic reticulum and Golgi. This AArf interacted with Sec23, a COPII vesicle coat component, in a GTP-dependent manner. These findings suggest certain AArfs are membrane-associating molecular switches with the functional potential to initiate organelle biogenesis, and the evolution of a functional coat could be the next critical step towards establishing eukaryotic cell architecture.
Collapse
Affiliation(s)
- Jing Zhu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, PR China
| | - Ruize Xie
- Key Laboratory of Polar Ecosystem and Climate Change, Ministry of Education, and School of Oceanography, Shanghai Jiao Tong University, Shanghai, PR China
| | - Qiaoying Ren
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, PR China
| | - Jiaming Zhou
- Key Laboratory of Polar Ecosystem and Climate Change, Ministry of Education, and School of Oceanography, Shanghai Jiao Tong University, Shanghai, PR China
| | - Chen Chen
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Meng-Xi Xie
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - You Zhou
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Yan Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Ningjing Liu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Jinchao Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, PR China
| | - Zhengwei Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Xipeng Liu
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China
| | - Wupeng Yan
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China.
| | - Qingqiu Gong
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China.
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, PR China.
| | - Liang Dong
- Key Laboratory of Polar Ecosystem and Climate Change, Ministry of Education, and School of Oceanography, Shanghai Jiao Tong University, Shanghai, PR China.
| | - Jinwei Zhu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, PR China.
- Department of Nephrology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Fengping Wang
- Key Laboratory of Polar Ecosystem and Climate Change, Ministry of Education, and School of Oceanography, Shanghai Jiao Tong University, Shanghai, PR China.
| | - Zhiping Xie
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, PR China.
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, PR China.
| |
Collapse
|
113
|
Charlanne L, Bertile F, Geffroy A, Hippauf L, Chery I, Zahn S, Guinet C, Piot E, Badaut J, Ancel A, Gilbert C, Bergouignan A. Ready to dive? Early constraints help juvenile southern elephant seals (Mirounga leonina) acclimatize to aquatic life. J Exp Biol 2025; 228:jeb249813. [PMID: 39925150 DOI: 10.1242/jeb.249813] [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: 11/06/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025]
Abstract
Breath-holding foraging implies different adaptations to limit oxygen (O2) depletion and maximize foraging time. Physiological adjustments can be mediated through O2 consumption, driven by muscle mitochondria, which can also produce reactive oxygen species during reoxygenation. Southern elephant seals spend months foraging at sea, diving for up to 1 h. Pups transition abruptly to aquatic life after a post-weaning period, during which they fast and progressively increase their activity, making this period critical for the development of an adaptive response to oxygen restriction and oxidative stress. We compared the functional capacity of a swimming muscle in 5 recently weaned and 6 adult female southern elephant seals. High-resolution respirometry was employed to examine muscle mitochondrial respiratory capacity and differences in protein and gene expression of the main regulatory pathways were determined using LC-MS/MS and RT-qPCR, respectively. Oxidative damage was measured in the plasma. We found that juveniles have higher mitochondrial coupling efficiency compared with adults, probably as a response to growth and significant physical activity reported during the post-weaning period. There were no differences in oxidative damage, but adults had a higher level of antioxidant defenses. Both hypoxia and oxidative response pathways appeared less activated in juveniles. This study highlights the differences in muscle metabolism and the likely adaptive response to hypoxia and oxidative stress between juvenile and adult south elephant seals. It also suggests that early constraints such as fasting, physical activity and short-term low O2 partial pressure exposure could contribute to immediate and long-term responses and help to acclimatize juveniles to aquatic life.
Collapse
Affiliation(s)
- Laura Charlanne
- Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Fabrice Bertile
- Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
- Proteomics French Infrastructure, FR2048, ProFI, 67000 Strasbourg, France
| | - Alexandre Geffroy
- Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
- Proteomics French Infrastructure, FR2048, ProFI, 67000 Strasbourg, France
| | - Lea Hippauf
- CNRS UMR 5536, Université de Bordeaux, 33076 Bordeaux, France
| | - Isabelle Chery
- Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Sandrine Zahn
- Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Christophe Guinet
- Centre d'Études Biologiques de Chizé, UMR 7372 CNRS/Université de La Rochelle, 79360 Villiers-en-Bois, France
| | - Erwan Piot
- CNRS UMR 5536, Université de Bordeaux, 33076 Bordeaux, France
- UMR 7179, CNRS/MNHN, Laboratoire MECADEV, 1 avenue du petit château, 91400 Brunoy, France
| | - Jérome Badaut
- CNRS UMR 5536, Université de Bordeaux, 33076 Bordeaux, France
| | - André Ancel
- Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - Caroline Gilbert
- UMR 7179, CNRS/MNHN, Laboratoire MECADEV, 1 avenue du petit château, 91400 Brunoy, France
- Ecole Nationale Vétérinaire d'Alfort, 7 avenue du Général de Gaulle, 94704 Maisons-Alfort, France
| | | |
Collapse
|
114
|
Gunawardana PBW, Gohil K, Moon KM, Foster LJ, Williams FJ. Proteomic Investigation of Neurotrophic trans-Banglene Reveals Potential Link to Iron Homeostasis. Mol Neurobiol 2025:10.1007/s12035-025-04772-1. [PMID: 40085355 DOI: 10.1007/s12035-025-04772-1] [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: 09/08/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025]
Abstract
In an effort to gain insight into cellular systems impacted by neurotrophic trans-banglene (t-BG), global proteomic profiling and Western blot analyses were employed. Expression level changes in response to t-BG treatment were compared to those observed with nerve growth factor (NGF), a natural neurotrophic protein and functional analog to t-BG. Findings from these studies did not point to direct interception of NGF/TrkA signaling by t-BG. Instead, significant alterations in iron-binding and iron-regulating proteins were observed. While total iron levels showed no change across all treatments, intracellular iron measurements and mitochondrial iron measurements demonstrated lower ferrous (Fe2+) ion levels in t-BG treated cells but not in NGF treated cells. These results highlight a potential connection between iron regulation and neurotrophic activity, a relationship which has, to date, not been well studied. These results are also notable given that iron dysregulation occurs in most neurodegenerative disease settings, and that iron has been shown to facilitate protein aggregation and apoptotic mechanisms.
Collapse
Affiliation(s)
| | - Khyati Gohil
- Department of Chemistry, University of Alberta, Edmonton, AB, Canada
| | - Kyung-Mee Moon
- Department of Biochemistry & Molecular Biology, University of British Colombia, Vancouver, Canada
| | - Leonard J Foster
- Department of Biochemistry & Molecular Biology, University of British Colombia, Vancouver, Canada
| | | |
Collapse
|
115
|
Raas Q, Haouy G, de Calbiac H, Pasho E, Marian A, Guerrera IC, Rosello M, Oeckl P, Del Bene F, Catanese A, Ciura S, Kabashi E. TBK1 is involved in programmed cell death and ALS-related pathways in novel zebrafish models. Cell Death Discov 2025; 11:98. [PMID: 40075110 PMCID: PMC11903655 DOI: 10.1038/s41420-025-02374-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/27/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Pathogenic mutations within the TBK1 gene leading to haploinsufficiency are causative of amyotrophic lateral sclerosis (ALS). This gene is linked to autophagy and inflammation, two cellular mechanisms reported to be dysregulated in ALS patients, although its functional role in the pathogenesis could involve other players. We targeted the TBK1 ortholog in zebrafish, an optimal vertebrate model for investigating genetic defects in neurological disorders. We generated zebrafish models with invalidating tbk1 mutations using CRISPR-Cas9 or tbk1 knockdown models using antisense morpholino oligonucleotide (AMO). The early motor phenotype of zebrafish injected with tbk1 AMO beginning at 2 days post fertilization (dpf) is associated with the degeneration of motor neurons. In parallel, CRISPR-induced tbk1 mutants exhibit impaired motor function beginning at 5 dpf and increased lethality beginning at 9 dpf. A metabolomic analysis showed an association between tbk1 loss and severe dysregulation of nicotinamide metabolism, and incubation with nicotinamide riboside rescued the motor behavior of tbk1 mutant zebrafish. Furthermore, a proteomic analysis revealed increased levels of inflammatory markers and dysregulation of programmed cell death pathways. Necroptosis appeared to be strongly activated in TBK1 fish, and larvae treated with the necroptosis inhibitor necrosulfonamide exhibited improved survival. Finally, a combined analysis of mutant zebrafish and TBK1-mutant human motor neurons revealed dysregulation of the KEGG pathway "ALS", with disrupted nuclear-cytoplasmic transport and increased expression of STAT1. These findings point toward a major role for necroptosis in the degenerative features and premature lethality observed in tbk1 mutant zebrafish. Overall, the novel tbk1-deficient zebrafish models offer a great opportunity to better understand the cascade of events leading from the loss of tbk1 expression to the onset of motor deficits, with involvement of a metabolic defect and increased cell death, and for the development of novel therapeutic avenues for ALS and related neuromuscular diseases.
Collapse
Affiliation(s)
- Quentin Raas
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Gregoire Haouy
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Hortense de Calbiac
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Elena Pasho
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Anca Marian
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Ida Chiara Guerrera
- Proteomics Platform 3P5-Necker, - Structure Fédérative de Recherche Necker, Inserm US24/CNRS UAR 3633, Université Paris Cité, 75015, Paris, France
| | - Marion Rosello
- Sorbonne Université, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Patrick Oeckl
- German Center for Neurodegenerative Diseases (DZNE) Ulm, Ulm, Germany
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Filippo Del Bene
- Sorbonne Université, INSERM U968, CNRS UMR 7210, Institut de la Vision, Paris, France
| | - Alberto Catanese
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
- Institute of Anatomy and Cell Biology, Ulm University School of Medicine, Ulm, Germany
| | - Sorana Ciura
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France
| | - Edor Kabashi
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, INSERM UMR 1163, Université Paris Cité, 75015, Paris, France.
| |
Collapse
|
116
|
Buri MV, Garavelli GY, Vigerelli H, de Souza MM, Maia Lobba AR, Ghidelli-Disse S, Chudzinski-Tavassi AM. cAmbly: A non-toxic cell-penetrating peptide derived from Amblyomin-X with targeted delivery to mitochondrial and cytoplasmic proteins. PLoS One 2025; 20:e0318119. [PMID: 40072951 PMCID: PMC11902296 DOI: 10.1371/journal.pone.0318119] [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: 12/21/2023] [Accepted: 01/10/2025] [Indexed: 03/14/2025] Open
Abstract
The effective delivery of drugs remains a major challenge in the development of new therapeutic molecules. Several strategies have been employed to address this issue, with cell-penetrating peptides (CPPs) standing out due to their ability to traverse cell membranes with minimal cytotoxicity and their relatively straightforward synthesis when conjugated with other molecules. However, while CPPs can successfully enter the cytoplasm, they often lack specificity for particular organelles, leaving target engagement to the drug itself. In this study, we present cAmbly, a novel CPP derived from the antitumoral protein Amblyomin-X. Our findings demonstrate that cAmbly efficiently internalizes into T98G cells within 30 min of incubation and preferentially colocalizes with mitochondria, exhibiting a clear affinity for mitochondrial proteins. These results suggest that cAmbly could serve as a promising delivery vehicle for mitochondria-targeted drugs, such as BCL-2 or OXPHOS modulators, which are commonly employed in cancer treatment. Furthermore, we identified the C-terminal of cAmbly as the optimal site for conjugating molecules intended for intracellular delivery.
Collapse
Affiliation(s)
- Marcus Vinicius Buri
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | | | - Hugo Vigerelli
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | | | - Aline Ramos Maia Lobba
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | | | | |
Collapse
|
117
|
Movassaghi CS, Sun J, Jiang Y, Turner N, Chang V, Chung N, Chen RJ, Browne EN, Lin C, Schweppe DK, Malaker SA, Meyer JG. Recent Advances in Mass Spectrometry-Based Bottom-Up Proteomics. Anal Chem 2025; 97:4728-4749. [PMID: 40000226 DOI: 10.1021/acs.analchem.4c06750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Mass spectrometry-based proteomics is about 35 years old, and recent progress appears to be speeding up across all subfields. In this review, we focus on advances over the last two years in select areas within bottom-up proteomics, including approaches to high-throughput experiments, data analysis using machine learning, drug discovery, glycoproteomics, extracellular vesicle proteomics, and structural proteomics.
Collapse
Affiliation(s)
- Cameron S Movassaghi
- Department of Computational Biomedicine, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
| | - Jie Sun
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Yuming Jiang
- Department of Computational Biomedicine, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
| | - Natalie Turner
- Departments of Molecular Medicine and Neurobiology, Scripps Research Institute, La Jolla, California 92037, United States
| | - Vincent Chang
- Department of Chemistry, Yale University, 275 Prospect Street, New Haven, Connecticut 06511, United States
| | - Nara Chung
- Department of Chemistry, Yale University, 275 Prospect Street, New Haven, Connecticut 06511, United States
| | - Ryan J Chen
- Department of Chemistry, Yale University, 275 Prospect Street, New Haven, Connecticut 06511, United States
| | - Elizabeth N Browne
- Department of Chemistry, Yale University, 275 Prospect Street, New Haven, Connecticut 06511, United States
| | - Chuwei Lin
- Department of Genome Sciences, University of Washington, Seattle, Washington 98105, United States
| | - Devin K Schweppe
- Department of Genome Sciences, University of Washington, Seattle, Washington 98105, United States
| | - Stacy A Malaker
- Department of Chemistry, Yale University, 275 Prospect Street, New Haven, Connecticut 06511, United States
| | - Jesse G Meyer
- Department of Computational Biomedicine, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
- Smidt Heart Institute, Cedars Sinai Medical Center, Los Angeles, California 90048, United States
| |
Collapse
|
118
|
Hu Y, Zhou T, Cai P, He Z. Neoantigens: new hope for cancer therapy. Front Oncol 2025; 15:1531592. [PMID: 40134605 PMCID: PMC11932895 DOI: 10.3389/fonc.2025.1531592] [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: 11/20/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
As research into tumour immunotherapy continues to accelerate, new frontiers are being revealed in the field of cancer treatment. A significant focus has been drawn to neoantigen-based personalised tumour vaccines, a pioneering immunotherapy. This approach involves the use of genetic mutations that are unique to tumor cells to custom-design personalized tumor vaccines. These vaccines elicit an immune response that is specifically directed at targeting and eliminating cancer cells. The incorporation of neoantigens, arising from mutations within tumor cells, confers a distinct advantage to personalized tumor vaccines in terms of precision and the mitigation of adverse effects. However, the intricate pathways from antigen presentation to the activation of tumor immunogenicity remain to be elucidated. This paper primarily delves into the origins and characteristics of neoantigens, and also neoantigen prediction, highlights existing screening methods, and addresses the limitations of current approaches. It is hoped that this review will act as a catalyst, accelerating the understanding of relevant knowledge and illuminating research hotspots for scientists poised to venture into neoantigen research.
Collapse
Affiliation(s)
- Yitong Hu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Tengda Zhou
- Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Ping Cai
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Zihao He
- Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
119
|
Rajagopal V, Seiler J, Nasa I, Cantarella S, Theiss J, Herget F, Kaifer B, Klostermann M, Will R, Schneider M, Helm D, König J, Zarnack K, Diederichs S, Kettenbach AN, Caudron-Herger M. An atlas of RNA-dependent proteins in cell division reveals the riboregulation of mitotic protein-protein interactions. Nat Commun 2025; 16:2325. [PMID: 40057470 PMCID: PMC11890761 DOI: 10.1038/s41467-025-57671-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/28/2025] [Indexed: 05/13/2025] Open
Abstract
Ribonucleoprotein complexes are dynamic assemblies of RNA with RNA-binding proteins, which modulate the fate of RNA. Inversely, RNA riboregulates the interactions and functions of the associated proteins. Dysregulation of ribonucleoprotein functions is linked to diseases such as cancer and neurological disorders. In dividing cells, RNA and RNA-binding proteins are present in mitotic structures, but their impact on cell division remains unclear. By applying the proteome-wide R-DeeP strategy to cells synchronized in mitosis versus interphase integrated with the RBP2GO knowledge, we provided an atlas of RNA-dependent proteins in cell division, accessible at R-DeeP3.dkfz.de. We uncovered AURKA, KIFC1 and TPX2 as unconventional RNA-binding proteins. KIFC1 was identified as a new substrate of AURKA, and new TPX2-interacting protein. Their pair-wise interactions were RNA dependent. In addition, RNA stimulated AURKA kinase activity and stabilized its conformation. In this work, we highlighted riboregulation of major mitotic factors as an additional complexity level of cell division.
Collapse
Affiliation(s)
- Varshni Rajagopal
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeanette Seiler
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Isha Nasa
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Simona Cantarella
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jana Theiss
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Franziska Herget
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bianca Kaifer
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melina Klostermann
- Buchmann Institute for Molecular Life Sciences, Frankfurt, Germany
- Department of Bioinformatics, University of Würzburg, Würzburg, Germany
| | - Rainer Will
- Cellular Tools Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martin Schneider
- Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dominic Helm
- Proteomics Core Facility, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julian König
- Institute of Molecular Biology (IMB), Mainz, Germany
- Theodor Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Kathi Zarnack
- Buchmann Institute for Molecular Life Sciences, Frankfurt, Germany
- Department of Bioinformatics, University of Würzburg, Würzburg, Germany
| | - Sven Diederichs
- Division of Cancer Research, Department of Thoracic Surgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center Freiburg, Freiburg, Germany.
| | - Arminja N Kettenbach
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
- Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA.
| | - Maïwen Caudron-Herger
- Research Group "RNA-Protein Complexes & Cell Proliferation", German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
120
|
Bond ST, King EJ, Walker SM, Yang C, Liu Y, Liu KH, Zhuang A, Jurrjens AW, Fang HA, Formosa LE, Nath AP, Carmona SR, Inouye M, Duong T, Huynh K, Meikle PJ, Crawford S, Ramm G, Elahee Doomun SN, de Souza DP, Rudler DL, Calkin AC, Filipovska A, Greening DW, Henstridge DC, Drew BG. Mitochondrial damage in muscle specific PolG mutant mice activates the integrated stress response and disrupts the mitochondrial folate cycle. Nat Commun 2025; 16:2338. [PMID: 40057508 PMCID: PMC11890779 DOI: 10.1038/s41467-025-57299-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 02/13/2025] [Indexed: 05/13/2025] Open
Abstract
During mitochondrial damage, information is relayed between the mitochondria and nucleus to coordinate precise responses to preserve cellular health. One such pathway is the mitochondrial integrated stress response (mtISR), which is known to be activated by mitochondrial DNA (mtDNA) damage. However, the causal molecular signals responsible for activation of the mtISR remain mostly unknown. A gene often associated with mtDNA mutations/deletions is Polg1, which encodes the mitochondrial DNA Polymerase γ (PolG). Here, we describe an inducible, tissue specific model of PolG mutation, which in muscle specific animals leads to rapid development of mitochondrial dysfunction and muscular degeneration in male animals from ~5 months of age. Detailed molecular profiling demonstrated robust activation of the mtISR in muscles from these animals. This was accompanied by striking alterations to enzymes in the mitochondrial folate cycle that was likely driven by a specific depletion in the folate cycle metabolite 5,10 methenyl-THF, strongly implying imbalanced folate intermediates as a previously unrecognised pathology linking the mtISR and mitochondrial disease.
Collapse
Affiliation(s)
- Simon T Bond
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| | - Emily J King
- Baker Heart & Diabetes Institute, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| | - Shannen M Walker
- Baker Heart & Diabetes Institute, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| | | | - Yingying Liu
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Kevin H Liu
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Aowen Zhuang
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Aaron W Jurrjens
- Baker Heart & Diabetes Institute, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
| | - Haoyun A Fang
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Luke E Formosa
- Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Artika P Nath
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | | | | | - Thy Duong
- Baker Heart & Diabetes Institute, Melbourne, Australia
| | - Kevin Huynh
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Peter J Meikle
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | - Simon Crawford
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Melbourne, Australia
| | - Georg Ramm
- Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
- Monash Ramaciotti Centre for Cryo Electron Microscopy, Monash University, Melbourne, Australia
| | | | | | - Danielle L Rudler
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Western Australia, Nedlands, Australia
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Western Australia, Nedlands, Australia
| | - Anna C Calkin
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| | - Aleksandra Filipovska
- ARC Centre of Excellence in Synthetic Biology, QEII Medical Centre, Western Australia, Nedlands, Australia
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, 15 Hospital Avenue, Western Australia, Nedlands, Australia
| | - David W Greening
- Baker Heart & Diabetes Institute, Melbourne, Australia
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
- School of Translational Medicine, Monash University, Melbourne, Australia
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia
| | - Darren C Henstridge
- Baker Heart & Diabetes Institute, Melbourne, Australia
- School of Health Sciences, University of Tasmania, Launceston, Australia
| | - Brian G Drew
- Baker Heart & Diabetes Institute, Melbourne, Australia.
- Baker Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia.
- School of Translational Medicine, Monash University, Melbourne, Australia.
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Australia.
| |
Collapse
|
121
|
Patocka T, Gupta S, Mastroleo F, Leys N, Matroule JY, Van Houdt R. Time-resolved proteomic profiling of Cupriavidus metallidurans CH34 in the copper-induced viable-but-nonculturable state. Metallomics 2025; 17:mfaf007. [PMID: 39963715 PMCID: PMC11886801 DOI: 10.1093/mtomcs/mfaf007] [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: 10/02/2024] [Accepted: 02/14/2025] [Indexed: 03/08/2025]
Abstract
Copper-based materials are actively explored for their potential as antimicrobial agents. However, recent studies show that sublethal concentrations of Cu ions can induce the viable-but-nonculturable (VBNC) cell state in certain bacteria, hampering contamination control, and monitoring. In this study we contribute to the unravelling of this largely enigmatic phenomenon by determining the time-resolved proteome of Cu-treated Cupriavidus metallidurans CH34 during VBNC induction and resuscitation. High-throughput quantitative liquid chromatography tandem mass spectrometry (LC-MS/MS) analysis was performed at multiple sample time points, revealing the cellular adaptations that trigger VBNC formation and the characteristic spontaneous recovery of culturability. Entry into the VBNC state correlated with a widespread response to oxidative stress as well as downregulated pyruvate metabolism. The expression of specific metal resistance determinants changed with Cu exposure time and culminated in the strong upregulation of proteins linked to periplasmic Cu ion detoxification during the resuscitation phase. We suggest that this delayed induction of Cu resistance proteins is paralleled by the gradual reconstitution of energy reserves through metabolization of intracellular polyhydroxybutyrate, as supported by flow cytometric fluorescence measurements. Furthermore, Cu-treated cells showed upregulation of several motility and chemotaxis proteins, and increased cell motility was observed phenotypically. Our results reveal a highly dynamic proteomic response, provide fundamental insights into the VBNC state and emphasize the advantages of time-resolved proteomic analysis.
Collapse
Affiliation(s)
- Timothej Patocka
- Microbiology Unit, Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
- Research Unit in Biology of Microorganisms, University of Namur, Namur, Belgium
| | - Surya Gupta
- Microbiology Unit, Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Felice Mastroleo
- Microbiology Unit, Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Natalie Leys
- Microbiology Unit, Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Jean-Yves Matroule
- Research Unit in Biology of Microorganisms, University of Namur, Namur, Belgium
| | - Rob Van Houdt
- Microbiology Unit, Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| |
Collapse
|
122
|
Ning D, Deng Y, Gao T, Yang Y, Chen G, Tian SZ, Zheng M. TF-chRDP: a method for simultaneously capturing transcription factor binding chromatin-associated RNA, DNA and protein. Front Cell Dev Biol 2025; 13:1561540. [PMID: 40123855 PMCID: PMC11925928 DOI: 10.3389/fcell.2025.1561540] [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: 01/16/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Transcription factors (TFs) play a crucial role in the regulation of gene expression and the structural organization of chromatin. They interact with proteins, RNA, and chromatin DNA to exert their functions. Therefore, an efficient and straightforward experimental approach that simultaneously captures the interactions of transcription factors with DNA, RNA, and proteins is essential for studying these regulatory proteins. In this study, we developed a novel method, TF-chRDP (Transcription Factor binding Chromatin-associated RNA, DNA, and Protein), which allows for the concurrent capture of these biomolecules in a single experiment. We enriched chromatin complexes using specific antibodies and divided the chromatin into three fractions: one for DNA library preparation to analyze the genomic binding sites of transcription factors, another for RNA library preparation to investigate the RNA associated with transcription factor binding, and the third for proteomic analysis to identify protein cofactors interacting with transcription factors. We applied this method to study the transcription factor p53 and its associated chromatin complexes. The results demonstrated high specificity in the enrichment of DNA, RNA and proteins. This method provides an efficient tool for simultaneously capturing chromatin-associated RNA, DNA and protein bound to specific TF, making it particularly useful for analyzing the role of protein-DNA-RNA complexes in transcriptional regulation.
Collapse
Affiliation(s)
- Duo Ning
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yuqing Deng
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Tong Gao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yang Yang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Gengzhan Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Simon Zhongyuan Tian
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Meizhen Zheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
123
|
Ferretti D, Kyriakidou P, Xiao J, Urazbakhtin S, De Nart C, Cox J. Isobaric Labeling Update in MaxQuant. J Proteome Res 2025; 24:1219-1229. [PMID: 39999392 PMCID: PMC11894648 DOI: 10.1021/acs.jproteome.4c00869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/02/2024] [Accepted: 02/20/2025] [Indexed: 02/27/2025]
Abstract
We present an update of the MaxQuant software for isobaric labeling data and evaluate its performance on benchmark data sets. Impurity correction factors can be applied to labels mixing C- and N-type reporter ions such as TMT Pro. Application to a single-cell multispecies mixture benchmark shows the high accuracy of the impurity-corrected results. TMT data recorded with FAIMS separation can be analyzed directly in MaxQuant without splitting the raw data into separate files per FAIMS voltage. Weighted median normalization is applied to several data sets, including large-scale human body atlas data. In the benchmark data sets, the weighted median normalization either removes or strongly reduces the batch effects between different TMT plexes and results in clustering by biology. In data sets including reference channels, we find that weighted median normalization performs as well or better when the reference channels are ignored and only the sample channel intensities are used, suggesting that the measurement of reference channels is unnecessary when using weighted median normalization in MaxQuant. We demonstrate that MaxQuant including the weighted median normalization performs well on multinotch MS3 data, as well as on phosphorylation data. MaxQuant is freely available for any purpose and can be downloaded from https://www.maxquant.org/.
Collapse
Affiliation(s)
| | | | | | - Shamil Urazbakhtin
- Computational Systems Biochemistry
Research Group, Max Planck Institute of
Biochemistry, Am Klopferspitz
18, Martinsried 82152, Germany
| | - Carlo De Nart
- Computational Systems Biochemistry
Research Group, Max Planck Institute of
Biochemistry, Am Klopferspitz
18, Martinsried 82152, Germany
| | - Jürgen Cox
- Computational Systems Biochemistry
Research Group, Max Planck Institute of
Biochemistry, Am Klopferspitz
18, Martinsried 82152, Germany
| |
Collapse
|
124
|
Grover CE, Jareczek JJ, Swaminathan S, Lee Y, Howell AH, Rani H, Arick MA, Leach AG, Miller ER, Yang P, Hu G, Xiong X, Mallery EL, Peterson DG, Xie J, Haigler CH, Zabotina OA, Szymanski DB, Wendel JF. A high-resolution model of gene expression during Gossypium hirsutum (cotton) fiber development. BMC Genomics 2025; 26:221. [PMID: 40050725 PMCID: PMC11884195 DOI: 10.1186/s12864-025-11360-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: 10/18/2024] [Accepted: 02/11/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Cotton fiber development relies on complex and intricate biological processes to transform newly differentiated fiber initials into the mature, extravagantly elongated cellulosic cells that are the foundation of this economically important cash crop. Here we extend previous research into cotton fiber development by employing controlled conditions to minimize variability and utilizing time-series sampling and analyses to capture daily transcriptomic changes from early elongation through the early stages of secondary wall synthesis (6 to 24 days post anthesis; DPA). RESULTS A majority of genes are expressed in fiber, largely partitioned into two major coexpression modules that represent genes whose expression generally increases or decreases during development. Differential gene expression reveals a massive transcriptomic shift between 16 and 17 DPA, corresponding to the onset of the transition phase that leads to secondary wall synthesis. Subtle gene expression changes are captured by the daily sampling, which are discussed in the context of fiber development. Coexpression and gene regulatory networks are constructed and associated with phenotypic aspects of fiber development, including turgor and cellulose production. Key genes are considered in the broader context of plant secondary wall synthesis, noting their known and putative roles in cotton fiber development. CONCLUSIONS The analyses presented here highlight the importance of fine-scale temporal sampling on understanding developmental processes and offer insight into genes and regulatory networks that may be important in conferring the unique fiber phenotype.
Collapse
Affiliation(s)
- Corrinne E Grover
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, 50011, USA.
| | - Josef J Jareczek
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, 50011, USA
- Present address: Bellarmine University, Louisville, KY, USA
| | - Sivakumar Swaminathan
- Roy J Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, 50011, USA
| | - Youngwoo Lee
- Department of Botany and Plant Pathology, Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
| | - Alexander H Howell
- Department of Botany and Plant Pathology, Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
| | - Heena Rani
- Department of Botany and Plant Pathology, Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
- Present address: USDA-ARS, Cereal Crops Research Unit, Madison, WI, 53726, USA
| | - Mark A Arick
- Institute for Genomics, Biocomputing & Biotechnology, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Alexis G Leach
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, 50011, USA
- Present address: Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Emma R Miller
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, 50011, USA
| | - Pengcheng Yang
- Department of Statistics, Purdue University, West Lafayette, IN, 47907, USA
| | - Guanjing Hu
- State Key Laboratory of Cotton Biology, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China
| | - Xianpeng Xiong
- State Key Laboratory of Cotton Biology, Institute of Cotton Research, Chinese Academy of Agricultural Sciences, Anyang, 455000, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, China
| | - Eileen L Mallery
- Department of Botany and Plant Pathology, Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
| | - Daniel G Peterson
- Institute for Genomics, Biocomputing & Biotechnology, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Jun Xie
- Department of Statistics, Purdue University, West Lafayette, IN, 47907, USA
| | - Candace H Haigler
- Department of Crop & Soil Sciences, North Carolina State University, Raleigh, NC, 27695, USA
- Department of Plant & Microbial Biology, North Carolina State University, Raleigh, NC, 27695, USA
| | - Olga A Zabotina
- Roy J Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, 50011, USA
| | - Daniel B Szymanski
- Department of Botany and Plant Pathology, Center for Plant Biology, Purdue University, West Lafayette, IN, 47907, USA
| | - Jonathan F Wendel
- Department of Ecology, Evolution, and Organismal Biology, Iowa State University, Ames, IA, 50011, USA
| |
Collapse
|
125
|
Martinez-Seidel F, Suwanchaikasem P, Gentry-Torfer D, Rajarathinam Y, Ebert A, Erban A, Firmino A, Nie S, Leeming M, Williamson N, Roessner U, Kopka J, Boughton BA. Remodelled ribosomal populations synthesize a specific proteome in proliferating plant tissue during cold. Philos Trans R Soc Lond B Biol Sci 2025; 380:20230384. [PMID: 40045790 PMCID: PMC11883437 DOI: 10.1098/rstb.2023.0384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/21/2024] [Accepted: 09/11/2024] [Indexed: 03/09/2025] Open
Abstract
Plant acclimation occurs through system-wide mechanisms that include proteome shifts, some of which occur at the level of protein synthesis. All proteins are synthesized by ribosomes. Rather than being monolithic, transcript-to-protein translation machines, ribosomes can be selective and cause proteome shifts. In this study, we use apical root meristems of germinating seedlings of the monocotyledonous plant barley as a model to examine changes in protein abundance and synthesis during cold acclimation. We measured metabolic and physiological parameters that allowed us to compare protein synthesis in the cold to optimal rearing temperatures. We demonstrated that the synthesis and assembly of ribosomal proteins are independent processes in root proliferative tissue. We report the synthesis and accumulation of various macromolecular complexes and propose how ribosome compositional shifts may be associated with functional proteome changes that are part of successful cold acclimation. Our study indicates that translation initiation is limiting during cold acclimation while the ribosome population is remodelled. The distribution of the triggered ribosomal protein heterogeneity suggests that altered compositions may confer 60S subunits selective association capabilities towards translation initiation complexes. To what extent selective translation depends on heterogeneous ribo-proteome compositions in barley proliferative root tissue remains a yet unresolved question.This article is part of the discussion meeting issue 'Ribosome diversity and its impact on protein synthesis, development and disease'.
Collapse
Affiliation(s)
- Federico Martinez-Seidel
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, CO, USA
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, CO, USA
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Pipob Suwanchaikasem
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Dione Gentry-Torfer
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Yogeswari Rajarathinam
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Alina Ebert
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Alexander Erban
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Alexandre Firmino
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Shuai Nie
- Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria, Australia
| | - Michael Leeming
- Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria, Australia
- School of Chemistry, The University of Melbourne, Parkville, Victoria, Australia
| | - Nicholas Williamson
- Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Ute Roessner
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
- Research School of Biology, The Australian National University, Acton, Australia
| | - Joachim Kopka
- Molecular Physiology Department, Max Planck Institute of Molecular Plant Physiology, Potsdam-Golm, Germany
| | - Berin A. Boughton
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
- La Trobe Institute of Sustainable Agriculture and Food, La Trobe University, Bundoora, Victoria3083, Australia
| |
Collapse
|
126
|
Ye Y, Fang Y, Engholm-Keller K, Bechshøft MR, Chatterton DEW, Sangild PT, Nguyen DN, Bering SB, Lund MN. Protein Digestibility and Anti-inflammatory Activity of Processed Whey Protein Ingredients for Infant Formula. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:5465-5476. [PMID: 39977279 DOI: 10.1021/acs.jafc.4c08785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Conventional whey protein concentrate (WPC) and gently processed skim-milk-derived WPC (SPC) undergo heat processing to ensure microbial safety before use in infant formula products. Heat treatment and storage induce protein structural changes, which may modulate digestibility and bioactivity. The objective of the study was to evaluate the effect of heat treatment and storage on the SPC ingredient by subjecting it to heat treatment (80 °C, 30 s) with or without an additional six-week storage at 37 °C (resulting in HT-SPC and HTS-SPC, respectively) and to compare these effects with a reference WPC ingredient. Reducible aggregates were present in HT-SPC, HTS-SPC, and WPC but not in the control SPC ingredient. As assessed in vitro, infant gastric digestion conditions had a limited hydrolytic effect on whey proteins, while significant protein hydrolysis occurred under infant intestinal conditions. WPC was more digestible than SPC, and additional heat treatment of SPC increased the protein digestibility. The digested protein ingredients exhibited similar anti-inflammatory activity (e.g., inhibition of the NFκB pathway in THP-1 macrophages in vitro). In conclusion, the SPC ingredient was less digestible, which was improved by heat treatment but with similar bioactivity as a conventional WPC ingredient.
Collapse
Affiliation(s)
- Yuhui Ye
- Department of Food Science, University of Copenhagen, 1958 Frederiksberg C, Denmark
| | - Yajing Fang
- Department of Food Science, University of Copenhagen, 1958 Frederiksberg C, Denmark
| | | | | | | | - Per T Sangild
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Dyrlægevej 68, 1870 Frederiksberg C, Denmark
- Department of Pediatrics, Odense University Hospital, 5000 Odense C, Denmark
- Department of Neonatology, Rigshospitalet, 2100 Copenhagen Ø, Denmark
| | - Duc Ninh Nguyen
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Dyrlægevej 68, 1870 Frederiksberg C, Denmark
| | - Stine B Bering
- Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, Dyrlægevej 68, 1870 Frederiksberg C, Denmark
| | - Marianne N Lund
- Department of Food Science, University of Copenhagen, 1958 Frederiksberg C, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3, 2200 Copenhagen N, Denmark
| |
Collapse
|
127
|
Thurow C, Pelizaeus AM, Mrozek P, Hoßbach BM, Budimir J, Schmitt K, Valerius O, Braus G, Gatz C. Redox-inactive CC-type glutaredoxins interfere with TGA transcription factor-dependent repression of target promoters in roots. THE PLANT CELL 2025; 37:koaf038. [PMID: 40053521 PMCID: PMC11887855 DOI: 10.1093/plcell/koaf038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025]
Abstract
Changes in nitrogen (N) availability in the soil trigger transcriptional responses in plants to optimize N acquisition, allocation, and remobilization. In roots of N-starved Arabidopsis (Arabidopsis thaliana) plants, transcriptional activation of genes encoding, for example, low-affinity nitrate transporters, depends on 4 related C-TERMINALLY ENCODED PEPTIDE DOWNSTREAM (CEPD) proteins, also known as ROXY6, ROXY7, ROXY8, and ROXY9. All 21 ROXYs found in A. thaliana interact with members of the TGACG-binding (TGA) family of transcription factors. Here, we demonstrate that 2 Clade I TGAs (TGA1, TGA4) serve as molecular links between CEPDs and their target promoters in roots. In the roxy6 roxy7 roxy8 roxy9 quadruple mutant (named cepd in this manuscript), transcriptional activation of N-starvation-inducible genes is impaired, most likely due to the association of Clade I TGAs with a repressive complex at their target promoters. In wild-type plants, this repressive complex is nonfunctional, and gene expression may be regulated by the N supply-regulated ratio of CEPDs over opposing ROXYs containing the TOPLESS-interacting ALWL motif. Although CEPDs resemble glutaredoxins with glutathione-dependent oxidoreductase activity, a ROXY9 variant with a mutation in the catalytic cysteine in its putative active site can confer wild-type-like regulation of target genes. This finding demonstrates that ROXY9 does not function through redox-dependent mechanisms.
Collapse
Affiliation(s)
- Corinna Thurow
- Albrecht-von-Haller-Institut für Pflanzenwissenschaften, Georg-August-Universität Göttingen, Julia-Lermontowa-Weg 3, D-37077 Göttingen, Germany
| | - Anja Maren Pelizaeus
- Albrecht-von-Haller-Institut für Pflanzenwissenschaften, Georg-August-Universität Göttingen, Julia-Lermontowa-Weg 3, D-37077 Göttingen, Germany
| | - Pascal Mrozek
- Albrecht-von-Haller-Institut für Pflanzenwissenschaften, Georg-August-Universität Göttingen, Julia-Lermontowa-Weg 3, D-37077 Göttingen, Germany
| | - Ben Moritz Hoßbach
- Albrecht-von-Haller-Institut für Pflanzenwissenschaften, Georg-August-Universität Göttingen, Julia-Lermontowa-Weg 3, D-37077 Göttingen, Germany
| | - Jelena Budimir
- Albrecht-von-Haller-Institut für Pflanzenwissenschaften, Georg-August-Universität Göttingen, Julia-Lermontowa-Weg 3, D-37077 Göttingen, Germany
| | - Kerstin Schmitt
- Institut für Mikrobiologie und Genetik, Serviceeinheit LCMS Proteinanalytik, Georg-August-Universität Göttingen, Grisebachstraße 8, D-37077 Göttingen, Germany
| | - Oliver Valerius
- Institut für Mikrobiologie und Genetik, Serviceeinheit LCMS Proteinanalytik, Georg-August-Universität Göttingen, Grisebachstraße 8, D-37077 Göttingen, Germany
| | - Gerhard Braus
- Institut für Mikrobiologie und Genetik, Serviceeinheit LCMS Proteinanalytik, Georg-August-Universität Göttingen, Grisebachstraße 8, D-37077 Göttingen, Germany
| | - Christiane Gatz
- Albrecht-von-Haller-Institut für Pflanzenwissenschaften, Georg-August-Universität Göttingen, Julia-Lermontowa-Weg 3, D-37077 Göttingen, Germany
| |
Collapse
|
128
|
Staniak M, Huang T, Figueroa-Navedo AM, Kohler D, Choi M, Hinkle T, Kleinheinz T, Blake R, Rose CM, Xu Y, Jean Beltran PM, Xue L, Bogdan M, Vitek O. Relative quantification of proteins and post-translational modifications in proteomic experiments with shared peptides: a weight-based approach. Bioinformatics 2025; 41:btaf046. [PMID: 39888862 PMCID: PMC11879648 DOI: 10.1093/bioinformatics/btaf046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 11/27/2024] [Accepted: 01/28/2025] [Indexed: 02/02/2025] Open
Abstract
MOTIVATION Bottom-up mass spectrometry-based proteomics studies changes in protein abundance and structure across conditions. Since the currency of these experiments are peptides, i.e. subsets of protein sequences that carry the quantitative information, conclusions at a different level must be computationally inferred. The inference is particularly challenging in situations where the peptides are shared by multiple proteins or post-translational modifications. While many approaches infer the underlying abundances from unique peptides, there is a need to distinguish the quantitative patterns when peptides are shared. RESULTS We propose a statistical approach for estimating protein abundances, as well as site occupancies of post-translational modifications, based on quantitative information from shared peptides. The approach treats the quantitative patterns of shared peptides as convex combinations of abundances of individual proteins or modification sites, and estimates the abundance of each source in a sample together with the weights of the combination. In simulation-based evaluations, the proposed approach improved the precision of estimated fold changes between conditions. We further demonstrated the practical utility of the approach in experiments with diverse biological objectives, ranging from protein degradation and thermal proteome stability, to changes in protein post-translational modifications. AVAILABILITY AND IMPLEMENTATION The approach is implemented in an open-source R package MSstatsWeightedSummary. The package is currently available at https://github.com/Vitek-Lab/MSstatsWeightedSummary (doi: 10.5281/zenodo.14662989). Code required to reproduce the results presented in this article can be found in a repository https://github.com/mstaniak/MWS_reproduction (doi: 10.5281/zenodo.14656053).
Collapse
Affiliation(s)
- Mateusz Staniak
- Faculty of Mathematics and Computer Science, University of Wrocław, Wrocław, 50-383, Poland
- Centre for Statistics, Hasselt University, Diepenbeek, 3590, Belgium
| | - Ting Huang
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, United States
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, United States
| | - Amanda M Figueroa-Navedo
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, United States
| | - Devon Kohler
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, United States
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, United States
| | - Meena Choi
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Trent Hinkle
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Tracy Kleinheinz
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Robert Blake
- Department of Biochemical and Cellular Pharmacology, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Christopher M Rose
- Department of Microchemistry, Proteomics, and Lipidomics, Genentech, Inc., South San Francisco, CA, 94080, United States
| | - Yingrong Xu
- Discovery Sciences, Pfizer Inc., Groton, CT, 06340, United States
| | - Pierre M Jean Beltran
- Machine Learning and Computational Sciences, Pfizer Inc., Cambridge, MA, 02139, United States
| | - Liang Xue
- Machine Learning and Computational Sciences, Pfizer Inc., Cambridge, MA, 02139, United States
| | - Małgorzata Bogdan
- Faculty of Mathematics and Computer Science, University of Wrocław, Wrocław, 50-383, Poland
| | - Olga Vitek
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, United States
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, United States
| |
Collapse
|
129
|
Luo S, Peng H, Shi Y, Cai J, Zhang S, Shao N, Li J. Integration of proteomics profiling data to facilitate discovery of cancer neoantigens: a survey. Brief Bioinform 2025; 26:bbaf087. [PMID: 40052441 PMCID: PMC11886573 DOI: 10.1093/bib/bbaf087] [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: 10/17/2024] [Revised: 12/29/2024] [Accepted: 02/19/2025] [Indexed: 03/10/2025] Open
Abstract
Cancer neoantigens are peptides that originate from alterations in the genome, transcriptome, or proteome. These peptides can elicit cancer-specific T-cell recognition, making them potential candidates for cancer vaccines. The rapid advancement of proteomics technology holds tremendous potential for identifying these neoantigens. Here, we provided an up-to-date survey about database-based search methods and de novo peptide sequencing approaches in proteomics, and we also compared these methods to recommend reliable analytical tools for neoantigen identification. Unlike previous surveys on mass spectrometry-based neoantigen discovery, this survey summarizes the key advancements in de novo peptide sequencing approaches that utilize artificial intelligence. From a comparative study on a dataset of the HepG2 cell line and nine mixed hepatocellular carcinoma proteomics samples, we demonstrated the potential of proteomics for the identification of cancer neoantigens and conducted comparisons of the existing methods to illustrate their limits. Understanding these limits, we suggested a novel workflow for neoantigen discovery as perspectives.
Collapse
Affiliation(s)
- Shifu Luo
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Hui Peng
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore
| | - Ying Shi
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
- School of Computer and Information Technology, Shanxi University, Taiyuan, 030006, Shanxi, China
| | - Jiaxin Cai
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| | - Songming Zhang
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| | - Ningyi Shao
- Faculty of Health Sciences, University of Macau, Taipa, Macao SAR 999078, China
| | - Jinyan Li
- Faculty of Computer Science and Control Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518107, Guangdong, China
| |
Collapse
|
130
|
Wolf van der Meer J, Larue A, van der Knaap JA, Chalkley GE, Sijm A, Beikmohammadi L, Kozhevnikova EN, van der Vaart A, Tilly BC, Bezstarosti K, Dekkers DHW, Doff WAS, van de Wetering-Tieleman PJ, Lanko K, Barakat TS, Allertz T, van Haren J, Demmers JAA, Atlasi Y, Verrijzer CP. Hao-Fountain syndrome protein USP7 controls neuronal differentiation via BCOR-ncPRC1.1. Genes Dev 2025; 39:401-422. [PMID: 39919828 PMCID: PMC11875088 DOI: 10.1101/gad.352272.124] [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: 09/04/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025]
Abstract
Pathogenic variants in the ubiquitin-specific protease 7 (USP7) gene cause a neurodevelopmental disorder called Hao-Fountain syndrome. However, it remains unclear which of USP7's pleiotropic functions are relevant for neurodevelopment. Here, we present a combination of quantitative proteomics, transcriptomics, and epigenomics to define the USP7 regulatory circuitry during neuronal differentiation. USP7 activity is required for the transcriptional programs that direct both the differentiation of embryonic stem cells into neural stem cells and the neuronal differentiation of SH-SY5Y neuroblastoma cells. USP7 controls the dosage of the Polycomb monubiquitylated histone H2A lysine 119 (H2AK119ub1) ubiquitin ligase complexes ncPRC1.1 and ncPRC1.6. Loss-of-function experiments revealed that BCOR-ncPRC1.1, but not ncPRC1.6, is a key effector of USP7 during neuronal differentiation. Indeed, BCOR-ncPRC1.1 mediates a major portion of USP7-dependent gene regulation during this process. Besides providing a detailed map of the USP7 regulome during neurodifferentiation, our results suggest that USP7- and ncPRC1.1-associated neurodevelopmental disorders involve dysregulation of a shared epigenetic network.
Collapse
Affiliation(s)
- Joyce Wolf van der Meer
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Axelle Larue
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, United Kingdom
| | - Jan A van der Knaap
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Gillian E Chalkley
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Ayestha Sijm
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Leila Beikmohammadi
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Elena N Kozhevnikova
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Aniek van der Vaart
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Ben C Tilly
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Karel Bezstarosti
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
- Proteomics Center, Erasmus MC University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Dick H W Dekkers
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
- Proteomics Center, Erasmus MC University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Wouter A S Doff
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
- Proteomics Center, Erasmus MC University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - P Jantine van de Wetering-Tieleman
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
- Proteomics Center, Erasmus MC University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Kristina Lanko
- Department of Clinical Genetics, Erasmus MC University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics, Erasmus MC University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Tim Allertz
- Department of Cell Biology, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Jeffrey van Haren
- Department of Cell Biology, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Jeroen A A Demmers
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands;
- Proteomics Center, Erasmus MC University Medical Center, 3025 GD Rotterdam, The Netherlands
| | - Yaser Atlasi
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast BT9 7AE, United Kingdom;
| | - C Peter Verrijzer
- Department of Biochemistry, Erasmus University Medical Center, 3025 GD Rotterdam, The Netherlands;
| |
Collapse
|
131
|
Bezawork-Geleta A, Devereux CJ, Keenan SN, Lou J, Cho E, Nie S, De Souza DP, Narayana VK, Siddall NA, Rodrigues CHM, Portelli S, Zheng T, Nim HT, Ramialison M, Hime GR, Dodd GT, Hinde E, Ascher DB, Stroud DA, Watt MJ. Proximity proteomics reveals a mechanism of fatty acid transfer at lipid droplet-mitochondria- endoplasmic reticulum contact sites. Nat Commun 2025; 16:2135. [PMID: 40032835 PMCID: PMC11876333 DOI: 10.1038/s41467-025-57405-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
Membrane contact sites between organelles are critical for the transfer of biomolecules. Lipid droplets store fatty acids and form contacts with mitochondria, which regulate fatty acid oxidation and adenosine triphosphate production. Protein compartmentalization at lipid droplet-mitochondria contact sites and their effects on biological processes are poorly described. Using proximity-dependent biotinylation methods, we identify 71 proteins at lipid droplet-mitochondria contact sites, including a multimeric complex containing extended synaptotagmin (ESYT) 1, ESYT2, and VAMP Associated Protein B and C (VAPB). High resolution imaging confirms localization of this complex at the interface of lipid droplet-mitochondria-endoplasmic reticulum where it likely transfers fatty acids to enable β-oxidation. Deletion of ESYT1, ESYT2 or VAPB limits lipid droplet-derived fatty acid oxidation, resulting in depletion of tricarboxylic acid cycle metabolites, remodeling of the cellular lipidome, and induction of lipotoxic stress. These findings were recapitulated in Esyt1 and Esyt2 deficient mice. Our study uncovers a fundamental mechanism that is required for lipid droplet-derived fatty acid oxidation and cellular lipid homeostasis, with implications for metabolic diseases and survival.
Collapse
Affiliation(s)
| | - Camille J Devereux
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Stacey N Keenan
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Jieqiong Lou
- School of Physics, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Ellie Cho
- Biological Optical Microscopy Platform (BOMP), The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility (MMSPF), Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - David P De Souza
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Vinod K Narayana
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, 3052, Australia
| | - Nicole A Siddall
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Carlos H M Rodrigues
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Stephanie Portelli
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Tenghao Zheng
- School of Biological Sciences, Monash University, Clayton, VIC, 3800, Australia
| | - Hieu T Nim
- Murdoch Children's Research Institute, reNEW Novo Nordisk Foundation for Stem Cell Medicine, Melbourne, VIC, 3052, Australia
| | - Mirana Ramialison
- Murdoch Children's Research Institute, reNEW Novo Nordisk Foundation for Stem Cell Medicine, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Gary R Hime
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Garron T Dodd
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Elizabeth Hinde
- School of Physics, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - David B Ascher
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD, 4072, Australia
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - David A Stroud
- Murdoch Children's Research Institute, reNEW Novo Nordisk Foundation for Stem Cell Medicine, Melbourne, VIC, 3052, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, VIC, 3052, Australia
- Victorian Clinical Genetics Services, Royal Children's Hospital, Melbourne, VIC, 3052, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
132
|
Ye Z, Sabatier P, van der Hoeven L, Lechner MY, Phlairaharn T, Guzman UH, Liu Z, Huang H, Huang M, Li X, Hartlmayr D, Izaguirre F, Seth A, Joshi HJ, Rodin S, Grinnemo KH, Hørning OB, Bekker-Jensen DB, Bache N, Olsen JV. Enhanced sensitivity and scalability with a Chip-Tip workflow enables deep single-cell proteomics. Nat Methods 2025; 22:499-509. [PMID: 39820750 PMCID: PMC11903336 DOI: 10.1038/s41592-024-02558-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025]
Abstract
Single-cell proteomics (SCP) promises to revolutionize biomedicine by providing an unparalleled view of the proteome in individual cells. Here, we present a high-sensitivity SCP workflow named Chip-Tip, identifying >5,000 proteins in individual HeLa cells. It also facilitated direct detection of post-translational modifications in single cells, making the need for specific post-translational modification-enrichment unnecessary. Our study demonstrates the feasibility of processing up to 120 label-free SCP samples per day. An optimized tissue dissociation buffer enabled effective single-cell disaggregation of drug-treated cancer cell spheroids, refining overall SCP analysis. Analyzing nondirected human-induced pluripotent stem cell differentiation, we consistently quantified stem cell markers OCT4 and SOX2 in human-induced pluripotent stem cells and lineage markers such as GATA4 (endoderm), HAND1 (mesoderm) and MAP2 (ectoderm) in different embryoid body cells. Our workflow sets a benchmark in SCP for sensitivity and throughput, with broad applications in basic biology and biomedicine for identification of cell type-specific markers and therapeutic targets.
Collapse
Affiliation(s)
- Zilu Ye
- 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.
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
| | - Pierre Sabatier
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
- Cardio-Thoracic Translational Medicine Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Leander van der Hoeven
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Maico Y Lechner
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Teeradon Phlairaharn
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Ulises H Guzman
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Zhen Liu
- 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
| | - Haoran Huang
- Thermo Fisher Scientific (China) Co. Ltd, Shanghai, China
| | - Min Huang
- Thermo Fisher Scientific (China) Co. Ltd, Shanghai, China
| | - Xiangjun Li
- Thermo Fisher Scientific (China) Co. Ltd, Shanghai, China
| | | | | | | | - Hiren J Joshi
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Sergey Rodin
- Cardio-Thoracic Translational Medicine Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Karl-Henrik Grinnemo
- Cardio-Thoracic Translational Medicine Laboratory, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | | | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
133
|
Phillips E, van Enk S, Kildgaard S, Schlue S, Göttmann M, Jennings V, Bethke F, Müller G, Herold‐Mende C, Pastor‐Flores D, Schneider M, Helm D, Ostenfeld Larsen T, Goidts V. Malformin C preferentially kills glioblastoma stem-like cells via concerted induction of proteotoxic stress and autophagic flux blockade. Mol Oncol 2025; 19:785-807. [PMID: 39462997 PMCID: PMC11887673 DOI: 10.1002/1878-0261.13756] [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: 05/22/2024] [Revised: 08/12/2024] [Accepted: 10/10/2024] [Indexed: 10/29/2024] Open
Abstract
Glioblastoma is a highly aggressive brain tumor for which there is no cure. The dire prognosis of this disease is largely attributable to a high level of heterogeneity, including the presence of a subpopulation of tumor-initiating glioblastoma stem-like cells (GSCs), which are refractory to chemo- and radiotherapy. Here, in an unbiased marine-derived fungal extract screen, together with bioguided dereplication based on high-resolution mass spectrometry, we identified malformin C to preferentially induce cell death in patient-derived GSCs and explore the potential of this cyclic peptide as a therapeutic agent for glioblastoma. Malformin C significantly reduced tumor growth in an in vivo xenograft model of glioblastoma. Using transcriptomics and chemoproteomics, we found that malformin C binds to many proteins, leading to their aggregation, and rapidly induces the unfolded protein response, including autophagy, in GSCs. Crucially, chemical inhibition of translation using cycloheximide rescued malformin C-induced cell death in GSCs, demonstrating that the proteotoxic effect of the compound is necessary for its cytotoxicity. At the same time, malformin C appears to accumulate in lysosomes, disrupting autophagic flux, and driving cells to death. Supporting this, malformin C synergizes with chloroquine, an inhibitor of autophagy. Strikingly, we observed that autophagic flux is differentially regulated in GSCs compared with normal astrocytes. The sensitivity of GSCs to malformin C highlights the relevance of proteostasis and autophagy as a therapeutic vulnerability in glioblastoma.
Collapse
Affiliation(s)
- Emma Phillips
- Junior Research Group “Brain Tumor Translational Targets”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Sizèd van Enk
- Junior Research Group “Brain Tumor Translational Targets”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Sara Kildgaard
- Department of Biotechnology and Biomedicine, Section for Microbial and Chemical Ecology, Natural Product DiscoveryTechnical University of DenmarkCopenhagenDenmark
| | - Silja Schlue
- Junior Research Group “Brain Tumor Translational Targets”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Mona Göttmann
- Junior Research Group “Brain Tumor Translational Targets”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Victoria Jennings
- Junior Research Group “Brain Tumor Translational Targets”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Frederic Bethke
- Junior Research Group “Brain Tumor Translational Targets”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Gabriele Müller
- Junior Research Group “Brain Tumor Translational Targets”German Cancer Research Center (DKFZ)HeidelbergGermany
| | - Christel Herold‐Mende
- Division of Neurosurgical Research, Department of NeurosurgeryUniversity Hospital HeidelbergGermany
| | - Daniel Pastor‐Flores
- Division of Redox RegulationGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Martin Schneider
- Proteomics Core FacilityGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Dominic Helm
- Proteomics Core FacilityGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Thomas Ostenfeld Larsen
- Department of Biotechnology and Biomedicine, Section for Microbial and Chemical Ecology, Natural Product DiscoveryTechnical University of DenmarkCopenhagenDenmark
| | - Violaine Goidts
- Junior Research Group “Brain Tumor Translational Targets”German Cancer Research Center (DKFZ)HeidelbergGermany
| |
Collapse
|
134
|
Zhang J, Wei L, Zhang H, Ma X, Sun Y, Li R, Zhang C, Cai X, Qiao J, Meng Q. Proteomic insights into nematode-trapping fungi Arthrobotrys oligospora after their response to chitin. J Vet Res 2025; 69:71-82. [PMID: 40144063 PMCID: PMC11936082 DOI: 10.2478/jvetres-2025-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/03/2025] [Indexed: 03/28/2025] Open
Abstract
Introduction Nematode-trapping fungi (NTFs) can produce various chitinases to degrade nematode body wall and eggshell chitin during predation. However, the regulatory mechanisms of their expression of chitinases still remain unclear. The primary objective of this study was to elucidate the differential protein profile of A. oligospora, an NTF, in response to chitin. Material and Methods Colloidal chitin was added to induce the culture of A. oligospora, and the phenotypic differences before and after induction were observed under inverted microscope. The differential proteins before and after mycelium induction were screened by liquid chromatography-tandem mass spectrometry. The differentially expressed chitinase was expressed in Pichia yeast, and the recombinant enzyme was incubated with Caenorhabditis elegans and its egg suspension to explore its biological activity. Results It was found that there was a significant acceleration in the mycelial growth post chitin interaction in A. oligospora. A total of 1,124 differentially expressed proteins (DEPs) were identified between the control group (AO-c) and the experimental group (AO-e), with 183 upregulated and 941 downregulated. Gene Ontology analysis revealed that the DEPs acted in various metabolic processes with catalysis and binding functions. Kyoto Encyclopedia of Genes and Genomes analysis associated these proteins primarily with signalling pathways related to glucose metabolism. Three chitinases were significantly modulated among DEPs. Moreover, enzymatic activity assays demonstrated that one of them effectively degraded C. elegans and its eggs. Conclusion These findings suggest that A. oligospora can significantly alter its protein expression profile in response to chitin, thereby facilitating its sugar metabolism and mycelial development. Our study provided new insights into the regulatory mechanisms of nematode predation in A. oligospora.
Collapse
Affiliation(s)
- Jiahua Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang832003, China
| | - Lixiang Wei
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang832003, China
| | - Huimei Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang832003, China
| | - Xixi Ma
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang832003, China
| | - Yansen Sun
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang832003, China
| | - Ruobing Li
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang832003, China
| | - Chengzhi Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang832003, China
| | - Xuepeng Cai
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu730046, China
| | - Jun Qiao
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang832003, China
| | - Qingling Meng
- College of Animal Science and Technology, Shihezi University, Shihezi, Xinjiang832003, China
| |
Collapse
|
135
|
Gęgotek A, Moniuszko-Malinowska A, Kruszewska E, Skrzydlewska E. Proteomic changes in serum of patients with Erythema migrans, Anaplasma phagocytophilum infection and co-infection. Microb Pathog 2025; 200:107337. [PMID: 39921043 DOI: 10.1016/j.micpath.2025.107337] [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: 09/26/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND According to the worldwide growing number of examined tick-borne diseases, the aim of this study was to evaluate the changes in proteomic profile of human serum induced by the development of Erythema migrans (EM), human granulocytic anaplasmosis (AP) and co-infection before/after antibiotic therapy. METHODS A proteomics approach based on SDS-PAGE/LC-MS/MS analysis was used to determine the proteins expression and 15d-PGE2 adducts level with albumin isolated from the serum of patients and sex/age-matched healthy donors. FINDINGS In the serum proteome of the patients with EM or/and AP, significant changes occurred in the expression of the same top 15 modified proteins; however, each protein level was modified in the different way in terms of comparing B. burgdorferi and A. phagocytophilum infections, as well as before and after therapy. In the case of co-infection, the differences in protein expression before and after therapy were significantly lower than in the monoinfection. Moreover, both EM and AP infections/co-infection significantly increased the albumin ability to create adducts with 15d-PGE2. The therapy partially reversed this property only in the case of a single infection, but this effect was not observed for therapy of co-infections. INTERPRETATION The results demonstrate how challenging is the treatment of the tick-borne co-infections and how important is further analysis of this subject. Individual differences are also observed in each of examined in this study diseases, which makes it more difficult to develop a common biomarker for each of the tick-borne diseases.
Collapse
Affiliation(s)
- Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540, Bialystok, Poland.
| | - Ewelina Kruszewska
- Department of Infectious Diseases and Neuroinfections, Medical University of Bialystok, Zurawia 14, 15-540, Bialystok, Poland.
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| |
Collapse
|
136
|
Dróżdż M, Zuvanov L, Sasikumar G, Bose D, Bruening F, Robles MS, Preußner M, Wahl M, Heyd F. Immediate early splicing controls translation in activated T-cells and is mediated by hnRNPC2 phosphorylation. EMBO J 2025; 44:1692-1723. [PMID: 39948410 PMCID: PMC11914300 DOI: 10.1038/s44318-025-00374-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 03/19/2025] Open
Abstract
The fast and transient induction of immediate early genes orchestrates the cellular response to various stimuli. These stimuli trigger phosphorylation cascades that promote immediate early gene transcription independent of de novo protein synthesis. Here we show that the same phosphorylation cascades also target the splicing machinery, inducing an analogous splicing switch that we call immediate early splicing (IES). We characterize hnRNPC2-controlled IES, which depends on the MEK-ERK pathway and the T cell-specific kinase PKCθ. This splicing switch mainly targets components of the translation machinery, such as mRNAs encoding ribosomal proteins and eIF5A. Inducing the eIF5A IES protein variant is by itself sufficient to reduce global translation, and consistently, we observe reduced de novo protein synthesis early after T cell activation. We suggest that immediate early splicing and the ensuing transient decrease in translation efficiency help to coordinate the extensive changes in gene expression during T cell activation. Together, these findings set a paradigm for fast and transient alternative splicing in the immediate cellular response to activation, and provide evidence for its functional relevance during T-cell stimulation.
Collapse
Affiliation(s)
- Mateusz Dróżdż
- Institute of Chemistry and Biochemistry, Laboratory of RNA Biochemistry, Freie Universität Berlin, Takustr. 6, 14195, Berlin, Germany
| | - Luíza Zuvanov
- Institute of Chemistry and Biochemistry, Laboratory of RNA Biochemistry, Freie Universität Berlin, Takustr. 6, 14195, Berlin, Germany
| | - Gopika Sasikumar
- Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Freie Universität Berlin, Takustr. 6, 14195, Berlin, Germany
| | - Debojit Bose
- Institute of Chemistry and Biochemistry, Laboratory of RNA Biochemistry, Freie Universität Berlin, Takustr. 6, 14195, Berlin, Germany
| | - Franziska Bruening
- Institute of Medical Psychology and Biomedical Center, Faculty of Medicine, LMU, Munich, Germany
| | - Maria S Robles
- Institute of Medical Psychology and Biomedical Center, Faculty of Medicine, LMU, Munich, Germany
| | - Marco Preußner
- Institute of Chemistry and Biochemistry, Laboratory of RNA Biochemistry, Freie Universität Berlin, Takustr. 6, 14195, Berlin, Germany
| | - Markus Wahl
- Institute of Chemistry and Biochemistry, Laboratory of Structural Biochemistry, Freie Universität Berlin, Takustr. 6, 14195, Berlin, Germany
| | - Florian Heyd
- Institute of Chemistry and Biochemistry, Laboratory of RNA Biochemistry, Freie Universität Berlin, Takustr. 6, 14195, Berlin, Germany.
| |
Collapse
|
137
|
Shi SM, Suh RJ, Shon DJ, Garcia FJ, Buff JK, Atkins M, Li L, Lu N, Sun B, Luo J, To NS, Cheung TH, McNerney MW, Heiman M, Bertozzi CR, Wyss-Coray T. Glycocalyx dysregulation impairs blood-brain barrier in ageing and disease. Nature 2025; 639:985-994. [PMID: 40011765 PMCID: PMC11946907 DOI: 10.1038/s41586-025-08589-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 01/03/2025] [Indexed: 02/28/2025]
Abstract
The blood-brain barrier (BBB) is highly specialized to protect the brain from harmful circulating factors in the blood and maintain brain homeostasis1,2. The brain endothelial glycocalyx layer, a carbohydrate-rich meshwork composed primarily of proteoglycans, glycoproteins and glycolipids that coats the BBB lumen, is a key structural component of the BBB3,4. This layer forms the first interface between the blood and brain vasculature, yet little is known about its composition and roles in supporting BBB function in homeostatic and diseased states. Here we find that the brain endothelial glycocalyx is highly dysregulated during ageing and neurodegenerative disease. We identify significant perturbation in an underexplored class of densely O-glycosylated proteins known as mucin-domain glycoproteins. We demonstrate that ageing- and disease-associated aberrations in brain endothelial mucin-domain glycoproteins lead to dysregulated BBB function and, in severe cases, brain haemorrhaging in mice. Finally, we demonstrate that we can improve BBB function and reduce neuroinflammation and cognitive deficits in aged mice by restoring core 1 mucin-type O-glycans to the brain endothelium using adeno-associated viruses. Cumulatively, our findings provide a detailed compositional and structural mapping of the ageing brain endothelial glycocalyx layer and reveal important consequences of ageing- and disease-associated glycocalyx dysregulation on BBB integrity and brain health.
Collapse
Affiliation(s)
- Sophia M Shi
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Ryan J Suh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - D Judy Shon
- Department of Chemistry, Stanford University, Stanford, CA, USA
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - Francisco J Garcia
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Josephine K Buff
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Micaiah Atkins
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA
| | - Lulin Li
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Nannan Lu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Bryan Sun
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| | - Ning-Sum To
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
| | - Tom H Cheung
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong, China
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Daniel and Mayce Yu Molecular Neuroscience Center, The Hong Kong University of Science and Technology, Hong Kong, China
| | - M Windy McNerney
- Department of Psychiatry, Stanford University School of Medicine, Stanford, CA, USA
- MIRECC, Department of Veterans Affairs, Palo Alto, CA, USA
| | - Myriam Heiman
- Picower Institute for Learning and Memory, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA, USA.
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA.
| | - Tony Wyss-Coray
- Stanford Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University School of Medicine, Stanford, CA, USA.
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA.
| |
Collapse
|
138
|
Dadson P, Honka MJ, Suomi T, Haridas PAN, Rokka A, Palani S, Goltseva E, Wang N, Roivainen A, Salminen P, James P, Olkkonen VM, Elo LL, Nuutila P. Proteomic profiling reveals alterations in metabolic and cellular pathways in severe obesity and following metabolic bariatric surgery. Am J Physiol Endocrinol Metab 2025; 328:E311-E324. [PMID: 39819027 DOI: 10.1152/ajpendo.00220.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/12/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
In this study, we investigated the impact of bariatric surgery on the adipose proteome to better understand the metabolic and cellular mechanisms underlying weight loss following the procedure. A total of 46 patients with severe obesity were included, with samples collected both before and after bariatric surgery. In addition, 15 healthy individuals without obesity who did not undergo surgery served as controls and were studied once. We utilized quantitative liquid chromatography-tandem mass spectrometry analysis to conduct a large-scale proteomic study on abdominal subcutaneous biopsies obtained from the study participants. Our proteomic profiling revealed that among the 2,254 compared proteins, 46 were upregulated and 34 were downregulated 6 months post surgery compared with baseline [false discovery rate (FDR) < 0.01]. We observed a downregulation of proteins associated with mitochondrial integrity, amino acid catabolism, and lipid metabolism in the patients with severe obesity compared with the controls. Bariatric surgery was associated with an upregulation in pathways related to mitochondrial function, protein synthesis, folding and trafficking, actin cytoskeleton regulation, and DNA binding and repair. These findings emphasize the significant changes in metabolic and cellular pathways following bariatric surgery, highlighting the potential mechanisms underlying the observed health improvements postbariatric surgery. The data provided alongside this paper will serve as a valuable resource for the development of targeted therapeutic strategies for obesity and related metabolic complications. ClinicalTrials.gov registration numbers: NCT00793143 (registered on 19 November 2008) (https://clinicaltrials.gov/ct2/show/NCT00793143) and NCT01373892 (registered on 15 June 2011) (https://clinicaltrials.gov/ct2/show/NCT01373892).NEW & NOTEWORTHY Our study investigates the effects of metabolic bariatric surgery on adipose tissue proteins, highlighting the mechanisms driving weight loss postsurgery. Through extensive proteomic analysis of adipose biopsies from patients with severe obesity pre- and postsurgery, alongside healthy subjects without obesity, we identified significant alterations in metabolic pathways. These findings provide insights into potential therapeutic targets for obesity-related complications.
Collapse
Affiliation(s)
- Prince Dadson
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Miikka-Juhani Honka
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Division of Information Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | - Anne Rokka
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | | | - Elena Goltseva
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Ning Wang
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Paulina Salminen
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Department of Surgery, University of Turku, Turku, Finland
- Division of Digestive Surgery and Urology, Turku University Hospital, Turku, Finland
| | - Peter James
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| |
Collapse
|
139
|
Ordon J, Logemann E, Maier LP, Lee T, Dahms E, Oosterwijk A, Flores-Uribe J, Miyauchi S, Paoli L, Stolze SC, Nakagami H, Felix G, Garrido-Oter R, Ma KW, Schulze-Lefert P. Conserved immunomodulation and variation in host association by Xanthomonadales commensals in Arabidopsis root microbiota. NATURE PLANTS 2025; 11:612-631. [PMID: 39972185 PMCID: PMC11928319 DOI: 10.1038/s41477-025-01918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 01/14/2025] [Indexed: 02/21/2025]
Abstract
Suppression of chronic Arabidopsis immune responses is a widespread but typically strain-specific trait across the major bacterial lineages of the plant microbiota. We show by phylogenetic analysis and in planta associations with representative strains that immunomodulation is a highly conserved, ancestral trait across Xanthomonadales, and preceded specialization of some of these bacteria as host-adapted pathogens. Rhodanobacter R179 activates immune responses, yet root transcriptomics suggest this commensal evades host immune perception upon prolonged association. R179 camouflage likely results from combined activities of two transporter complexes (dssAB) and the selective elimination of immunogenic peptides derived from all partners. The ability of R179 to mask itself and other commensals from the plant immune system is consistent with a convergence of distinct root transcriptomes triggered by immunosuppressive or non-suppressive synthetic microbiota upon R179 co-inoculation. Immunomodulation through dssAB provided R179 with a competitive advantage in synthetic communities in the root compartment. We propose that extensive immunomodulation by Xanthomonadales is related to their adaptation to terrestrial habitats and might have contributed to variation in strain-specific root association, which together accounts for their prominent role in plant microbiota establishment.
Collapse
Affiliation(s)
- Jana Ordon
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
- Department of Plant and Microbial Biology, University of Zurich, Zurich, Switzerland
| | - Elke Logemann
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Louis-Philippe Maier
- Center for Plant Molecular Biology, University Tuebingen, Tuebingen, Germany
- Department of Plant Molecular Biology, University of Lausanne, Lausanne, Switzerland
| | - Tak Lee
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Eik Dahms
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
- Regional Computing Centre, University of Cologne, Cologne, Germany
| | - Anniek Oosterwijk
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
- Laboratory of Plant Physiology, Wageningen University and Research, Wageningen, the Netherlands
| | - Jose Flores-Uribe
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Shingo Miyauchi
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
- Cluster of Excellence on Plant Sciences, Max Planck Institute for Plant Breeding Research, Cologne, Germany
- Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| | - Lucas Paoli
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zurich, Zurich, Switzerland
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Sara Christina Stolze
- Protein Mass Spectrometry Group, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Hirofumi Nakagami
- Protein Mass Spectrometry Group, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Georg Felix
- Center for Plant Molecular Biology, University Tuebingen, Tuebingen, Germany
| | - Ruben Garrido-Oter
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
- Cluster of Excellence on Plant Sciences, Max Planck Institute for Plant Breeding Research, Cologne, Germany
- Earlham Institute, Norwich Research Park, Norwich, UK
| | - Ka-Wai Ma
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany.
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan.
| | - Paul Schulze-Lefert
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany.
- Cluster of Excellence on Plant Sciences, Max Planck Institute for Plant Breeding Research, Cologne, Germany.
| |
Collapse
|
140
|
Min CW, Gupta R, Lee GH, Cho JH, Kim YJ, Wang Y, Jung KH, Kim ST. Integrative Proteomic and Phosphoproteomic Profiling Reveals the Salt-Responsive Mechanisms in Two Rice Varieties (Oryza Sativa subsp. Japonica and Indica). Proteomics 2025; 25:e202400251. [PMID: 39491529 DOI: 10.1002/pmic.202400251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/09/2024] [Accepted: 10/11/2024] [Indexed: 11/05/2024]
Abstract
Salinity stress induces ionic and osmotic imbalances in rice plants that in turn negatively affect the photosynthesis rate, resulting in growth retardation and yield penalty. Efforts have, therefore, been carried out to understand the mechanism of salt tolerance, however, the complexity of biological processes at proteome levels remains a major challenge. Here, we performed a comparative proteome and phosphoproteome profiling of microsome enriched fractions of salt-tolerant (cv. IR73; indica) and salt-susceptible (cv. Dongjin/DJ; japonica) rice varieties. This approach led to the identification of 5856 proteins, of which 473 and 484 proteins showed differential modulation between DJ and IR73 sample sets, respectively. The phosphoproteome analysis led to the identification of a total of 10,873 phosphopeptides of which 2929 and 3049 phosphopeptides showed significant differences in DJ and IR73 sample sets, respectively. The integration of proteome and phosphoproteome data showed activation of ABA and Ca2+ signaling components exclusively in the salt-tolerant variety IR73 in response to salinity stress. Taken together, our results highlight the changes at proteome and phosphoproteome levels and provide a mechanistic understanding of salinity stress tolerance in rice.
Collapse
Affiliation(s)
- Cheol Woo Min
- Department of Plant Bioscience, Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| | - Ravi Gupta
- College of General Education, Kookmin University, Seoul, Republic of Korea
| | - Gi Hyun Lee
- Department of Plant Bioscience, Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| | - Jun-Hyeon Cho
- Sangju Substation, National Institute of Crop Science, Rural Development Administration (RDA), Sangju, Republic of Korea
| | - Yu-Jin Kim
- Department of Life Science and Environmental Biochemistry, Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| | - Yiming Wang
- Key Laboratory of Integrated Management of Crop Disease and Pests, Department of Plant Pathology, Ministry of Education, Nanjing Agricultural University, Nanjing, China
| | - Ki-Hong Jung
- Graduate School of Biotechnology and Crop Biotech Institute, Kyung Hee University, Yongin, Republic of Korea
| | - Sun Tae Kim
- Department of Plant Bioscience, Life and Industry Convergence Research Institute, Pusan National University, Miryang, Republic of Korea
| |
Collapse
|
141
|
Zhao Q, Jing Y, Jiang X, Zhang X, Liu F, Huang H, Zhang Z, Wang H, Sun S, Ma S, Zhang W, Yu Y, Fu X, Zhao G, Qu J, Wang S, Liu GH. SIRT5 safeguards against primate skeletal muscle ageing via desuccinylation of TBK1. Nat Metab 2025; 7:556-573. [PMID: 40087407 DOI: 10.1038/s42255-025-01235-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 02/06/2025] [Indexed: 03/17/2025]
Abstract
Ageing-induced skeletal muscle deterioration contributes to sarcopenia and frailty, adversely impacting the quality of life in the elderly. However, the molecular mechanisms behind primate skeletal muscle ageing remain largely unexplored. Here, we show that SIRT5 expression is reduced in aged primate skeletal muscles from both genders. SIRT5 deficiency in human myotubes hastens cellular senescence and intensifies inflammation. Mechanistically, we demonstrate that TBK1 is a natural substrate for SIRT5. SIRT5 desuccinylates TBK1 at lysine 137, which leads to TBK1 dephosphorylation and the suppression of the downstream inflammatory pathway. Using SIRT5 lentiviral vectors for skeletal muscle gene therapy in male mice enhances physical performance and alleviates age-related muscle dysfunction. This study sheds light on the molecular underpinnings of skeletal muscle ageing and presents the SIRT5-TBK1 pathway as a promising target for combating age-related skeletal muscle degeneration.
Collapse
Affiliation(s)
- Qian Zhao
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Ying Jing
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiaoyu Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xin Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
| | - Feifei Liu
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Haoyan Huang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China
- Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhihua Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
| | - Haijun Wang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
| | - Shuhui Sun
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shuai Ma
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Aging Biomarker Consortium (ABC), Beijing, China
| | - Weiqi Zhang
- University of Chinese Academy of Sciences, Beijing, China
- Aging Biomarker Consortium (ABC), Beijing, China
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics and China National Center for Bioinformation, Chinese Academy of Sciences, Beijing, China
- Sino-Danish College, Sino-Danish Center for Education and Research, University of Chinese Academy of Sciences, Beijing, China
| | - Yang Yu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Xiaobing Fu
- Tissue Repair and Regeneration Research Center, Medical Innovation Department, PLA General Hospital and Medical College, Beijing, China
| | - Guoguang Zhao
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
- National Medical Center for Neurological Diseases, Beijing, China
- Beijing Municipal Geriatric Medical Research Center, Beijing, China
| | - Jing Qu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Aging Biomarker Consortium (ABC), Beijing, China.
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China.
- Aging Translational Medicine Center, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium (ABC), Beijing, China.
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Aging Biomarker Consortium (ABC), Beijing, China.
| |
Collapse
|
142
|
Wang H, Syed AA, Krijgsveld J, Sigismondo G. Isolation of Proteins on Chromatin Reveals Signaling Pathway-Dependent Alterations in the DNA-Bound Proteome. Mol Cell Proteomics 2025; 24:100908. [PMID: 39842777 PMCID: PMC11889358 DOI: 10.1016/j.mcpro.2025.100908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 01/03/2025] [Accepted: 01/17/2025] [Indexed: 01/24/2025] Open
Abstract
Signaling pathways often convergence on transcription factors and other DNA-binding proteins that regulate chromatin structure and gene expression, thereby governing a broad range of essential cellular functions. However, the repertoire of DNA-binding proteins is incompletely understood even for the best-characterized pathways. Here, we optimized a strategy for the isolation of Proteins on Chromatin (iPOC) exploiting tagged nucleoside analogs to label the DNA and capture associated proteins, thus enabling the comprehensive, sensitive, and unbiased characterization of the DNA-bound proteome. We then applied iPOC to investigate chromatome changes upon perturbation of the cancer-relevant PI3K-AKT-mTOR pathway. Our results show distinct dynamics of the DNA-bound proteome upon selective inhibition of PI3K, AKT, or mTOR, and we provide evidence how this signaling cascade regulates the DNA-bound status of SUZ12, thereby modulating H3K27me3 levels. Collectively, iPOC is a powerful approach to study the composition of the DNA-bound proteome operating downstream of signaling cascades, thereby both expanding our knowledge of the mechanism of action of the pathway and unveiling novel chromatin modulators that can potentially be targeted pharmacologically.
Collapse
Affiliation(s)
- Huiyu Wang
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Azmal Ali Syed
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeroen Krijgsveld
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty, Heidelberg University, Heidelberg, Germany.
| | - Gianluca Sigismondo
- Division of Proteomics of Stem Cell and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany; Medical Faculty, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
143
|
Abele M, Soleymaniniya A, Bayer FP, Lomp N, Doll E, Meng C, Neuhaus K, Scherer S, Wenning M, Wantia N, Kuster B, Wilhelm M, Ludwig C. Proteomic Diversity in Bacteria: Insights and Implications for Bacterial Identification. Mol Cell Proteomics 2025; 24:100917. [PMID: 39880082 PMCID: PMC11919601 DOI: 10.1016/j.mcpro.2025.100917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 01/31/2025] Open
Abstract
Mass spectrometry-based proteomics has revolutionized bacterial identification and elucidated many molecular mechanisms underlying bacterial growth, community formation, and drug resistance. However, most research has been focused on a few model bacteria, overlooking bacterial diversity. In this study, we present the most extensive bacterial proteomic resource to date, covering 303 species, 119 genera, and five phyla with over 636,000 unique expressed proteins, confirming the existence of over 38,700 hypothetical proteins. Accessible via the public resource ProteomicsDB, this dataset enables quantitative exploration of proteins within and across species. Additionally, we developed MS2Bac, a bacterial identification algorithm that queries NCBI's bacterial proteome space in two iterations. MS2Bac achieved over 99% species-level and 89% strain-level accuracy, surpassing methods like MALDI-TOF and FTIR, as demonstrated with food-derived bacterial isolates. MS2Bac also effectively identified bacteria in clinical samples, highlighting the potential of MS-based proteomics as a routine diagnostic tool.
Collapse
Affiliation(s)
- Miriam Abele
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), TUM School of Life Sciences, Technical University of Munich, Freising, Germany; Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Armin Soleymaniniya
- Computational Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Florian P Bayer
- Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Nina Lomp
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Etienne Doll
- Research Department Molecular Life Sciences, TUM School of Life Sciences, Freising, Germany
| | - Chen Meng
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome, ZIEL Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Siegfried Scherer
- Research Department Molecular Life Sciences, TUM School of Life Sciences, Freising, Germany
| | - Mareike Wenning
- Bavarian Health and Food Safety Authority, Unit for Food Microbiology and Hygiene, Oberschleißheim, Germany
| | - Nina Wantia
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, TUM School of Medicine and Health Department Preclinical Medicine, Technical University of Munich, Munich, Germany
| | - Bernhard Kuster
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), TUM School of Life Sciences, Technical University of Munich, Freising, Germany; Chair of Proteomics and Bioanalytics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; Munich Data Science Institute (MDSI), Technical University of Munich, Garching, Germany
| | - Mathias Wilhelm
- Computational Mass Spectrometry, TUM School of Life Sciences, Technical University of Munich, Freising, Germany; Munich Data Science Institute (MDSI), Technical University of Munich, Garching, Germany
| | - Christina Ludwig
- Bavarian Center for Biomolecular Mass Spectrometry (BayBioMS), TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
144
|
Lim B, Kamal A, Gomez Ramos B, Adrian Segarra JM, Ibarra IL, Dignas L, Kindinger T, Volz K, Rahbari M, Rahbari N, Poisel E, Kafetzopoulou K, Böse L, Breinig M, Heide D, Gallage S, Barragan Avila JE, Wiethoff H, Berest I, Schnabellehner S, Schneider M, Becker J, Helm D, Grimm D, Mäkinen T, Tschaharganeh DF, Heikenwalder M, Zaugg JB, Mall M. Active repression of cell fate plasticity by PROX1 safeguards hepatocyte identity and prevents liver tumorigenesis. Nat Genet 2025; 57:668-679. [PMID: 39948437 PMCID: PMC11906372 DOI: 10.1038/s41588-025-02081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/08/2025] [Indexed: 02/20/2025]
Abstract
Cell fate plasticity enables development, yet unlocked plasticity is a cancer hallmark. While transcription master regulators induce lineage-specific genes to restrict plasticity, it remains unclear whether plasticity is actively suppressed by lineage-specific repressors. Here we computationally predict so-called safeguard repressors for 18 cell types that block phenotypic plasticity lifelong. We validated hepatocyte-specific candidates using reprogramming, revealing that prospero homeobox protein 1 (PROX1) enhanced hepatocyte identity by direct repression of alternative fate master regulators. In mice, Prox1 was required for efficient hepatocyte regeneration after injury and was sufficient to prevent liver tumorigenesis. In line with patient data, Prox1 depletion caused hepatocyte fate loss in vivo and enabled the transition of hepatocellular carcinoma to cholangiocarcinoma. Conversely, overexpression promoted cholangiocarcinoma to hepatocellular carcinoma transdifferentiation. Our findings provide evidence for PROX1 as a hepatocyte-specific safeguard and support a model where cell-type-specific repressors actively suppress plasticity throughout life to safeguard lineage identity and thus prevent disease.
Collapse
Affiliation(s)
- Bryce Lim
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Aryan Kamal
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Borja Gomez Ramos
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Juan M Adrian Segarra
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ignacio L Ibarra
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Lennart Dignas
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tim Kindinger
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kai Volz
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Mohammad Rahbari
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
- Department of Surgery, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Nuh Rahbari
- Department of Surgery, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of General and Visceral Surgery, University of Ulm, Ulm, Germany
| | - Eric Poisel
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kanela Kafetzopoulou
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lio Böse
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Marco Breinig
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Danijela Heide
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
| | - Suchira Gallage
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
- Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Faculty of Medicine, University Tuebingen, Tübingen, Germany
| | | | - Hendrik Wiethoff
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Ivan Berest
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Sarah Schnabellehner
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Jonas Becker
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, Heidelberg University, Center for Integrative Infectious Diseases Research (CIID), BioQuant, Heidelberg, Germany
| | - Dominic Helm
- Proteomics Core Facility, DKFZ, Heidelberg, Germany
| | - Dirk Grimm
- Department of Infectious Diseases/Virology, Section Viral Vector Technologies, Medical Faculty and Faculty of Engineering Sciences, Heidelberg University, Center for Integrative Infectious Diseases Research (CIID), BioQuant, Heidelberg, Germany
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg, Heidelberg, Germany
| | - Taija Mäkinen
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland
- Wihuri Research Institute, Helsinki, Finland
| | - Darjus F Tschaharganeh
- Cell Plasticity and Epigenetic Remodeling Helmholtz Group, DKFZ, Heidelberg, Germany
- Institute of Pathology, University Hospital, Heidelberg, Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, DKFZ, Heidelberg, Germany
- Institute for Interdisciplinary Research on Cancer Metabolism and Chronic Inflammation, M3-Research Center for Malignome, Metabolome and Microbiome, Faculty of Medicine, University Tuebingen, Tübingen, Germany
| | - Judith B Zaugg
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany.
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Moritz Mall
- Cell Fate Engineering and Disease Modeling Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- HITBR Hector Institute for Translational Brain Research gGmbH, Heidelberg, Germany.
- Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
145
|
Orchard SE. What have Data Standards ever done for us? Mol Cell Proteomics 2025:100933. [PMID: 40024375 DOI: 10.1016/j.mcpro.2025.100933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
The Human Proteome Organization (HUPO) Proteomics Standards Initiative (PSI) has been successfully developing guidelines, data formats, and controlled vocabularies for both the field of molecular interaction and that of mass spectrometry for more than 20 years. This review explores some of the ways that the proteomics community has benefitted from the development of community standards and takes a look at some of the tools and resources that have been improved or developed as a result of the work of the HUPO-PSI.
Collapse
Affiliation(s)
- S E Orchard
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton CB10 1SD, UK
| |
Collapse
|
146
|
Brinkley DM, Bertolli SK, Gallagher LA, Tan Y, de Silva MM, Brockman A, Zhang D, Peterson SB, Mougous JD. Pseudomonads coordinate innate defense against viruses and bacteria with a single regulatory system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640152. [PMID: 40060533 PMCID: PMC11888443 DOI: 10.1101/2025.02.26.640152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Bacterial cells live under the constant existential threats imposed by other bacteria and viruses. Their mechanisms for contending with these threats are well documented; however, the regulation of these diverse defense elements remains poorly understood. Here we show that bacteria can mount a genome-wide, coordinated, and highly effective immune response against bacterial and viral threats using a single regulatory pathway. Bioinformatic analyses revealed that Pseudomonas species broadly possess a specialized form of the Gac/Rsm regulatory pathway (GRP), which our prior work in Pseudomonas aeruginosa implicated in activating interbacterial antagonism defense mechanisms in response to neighbor cell death. Proteomic studies comparing GRP-activated and -inactivated strains derived from diverse Pseudomonas species showed that the pathway regulates a large and variable suite of factors implicated in defense against both bacterial and phage threats. Focusing on P. protegens, we identify profound phenotypic consequences of these factors against multiple forms of bacterial antagonism and several phage. Together, our results reveal that bacteria, like more complex organisms, couple danger sensing to the activation of an immune system with antibacterial and antiviral arms.
Collapse
Affiliation(s)
- David M Brinkley
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA, USA
| | | | - Larry A Gallagher
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Yongjun Tan
- Department of Biology, Saint Louis University, St. Louis, MO, USA
- Program of Bioinformatic and Computational Biology, Saint Louis University, St. Louis, MO USA
| | | | - Ainsley Brockman
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Dapeng Zhang
- Department of Biology, Saint Louis University, St. Louis, MO, USA
- Program of Bioinformatic and Computational Biology, Saint Louis University, St. Louis, MO USA
| | - S Brook Peterson
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Joseph D Mougous
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Microbial Interactions and Microbiome Center, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
147
|
Kloss LDF, Doellinger J, Gries A, Soler E, Lasch P, Heinz J. Proteomic insights into survival strategies of Escherichia coli in perchlorate-rich Martian brines. Sci Rep 2025; 15:6988. [PMID: 40011700 PMCID: PMC11865474 DOI: 10.1038/s41598-025-91562-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 02/21/2025] [Indexed: 02/28/2025] Open
Abstract
Brines, potentially formed by the deliquescence and freezing point depression of highly hygroscopic salts, such as perchlorates (ClO4-), may allow for the spatial and temporal stability of liquid water on present-day Mars. It is therefore of great interest to explore the microbial habitability of Martian brines, for which our current understanding is, however, still limited. Putative microbes growing in the perchlorate-rich Martian regolith may be harmed due to the induction of various stressors including osmotic, chaotropic, and oxidative stress. We adapted the model organism Escherichia coli to increasing sodium perchlorate concentrations and used a proteomic approach to characterize the adaptive phenotype. Separately, the microbe was adapted to elevated concentrations of sodium chloride and glycerol, which enabled us to distinguish perchlorate-specific adaptation mechanisms from those in response to osmotic, ion and water activity stress. We found that the perchlorate-specific stress response focused on pathways alleviating damage to nucleic acids, presumably caused by increased chaotropic and/or oxidative stress. The significant enrichments that have been found include DNA repair, RNA methylation and de novo inosine monophosphate (IMP) biosynthesis. Our study provides insights into the adaptive mechanisms necessary for microorganisms to survive under perchlorate stress, with implications for understanding the habitability of Martian brines.
Collapse
Affiliation(s)
- Lea D F Kloss
- Center for Astronomy and Astrophysics, RG Astrobiology, Technische Universität Berlin, Berlin, Germany
- Institute for Computer Science and Department of Biology, Heinrich Heine University, Düsseldorf, Germany
| | - Joerg Doellinger
- Centre for Biological Threats and Special Pathogens, Proteomics and Spectroscopy (ZBS6), Robert Koch-Institute, Berlin, Germany
| | - Anne Gries
- Center for Astronomy and Astrophysics, RG Astrobiology, Technische Universität Berlin, Berlin, Germany
| | - Elisa Soler
- Center for Astronomy and Astrophysics, RG Astrobiology, Technische Universität Berlin, Berlin, Germany
| | - Peter Lasch
- Centre for Biological Threats and Special Pathogens, Proteomics and Spectroscopy (ZBS6), Robert Koch-Institute, Berlin, Germany
| | - Jacob Heinz
- Center for Astronomy and Astrophysics, RG Astrobiology, Technische Universität Berlin, Berlin, Germany.
| |
Collapse
|
148
|
Brown JS, Lee MA, Vuong W, Loas A, Pentelute BL. pyBinder: Quantitation to Advance Affinity Selection-Mass Spectrometry. Anal Chem 2025; 97:3855-3863. [PMID: 39949296 DOI: 10.1021/acs.analchem.4c04445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
Affinity selection-mass spectrometry (AS-MS) is a ligand discovery platform that relies upon mass spectrometry to identify molecules bound to a biomolecular target. When utilized with large peptide libraries (108 members), AS-MS sample complexity can surpass the sequencing capacity of modern mass spectrometers, resulting in incomplete data, identification of few target-specific ligands, and/or incomplete sequencing. To address this challenge, we introduce pyBinder to perform quantitation on AS-MS data to process primary MS1 data and develop two scores to rank the peptides from the integration of their peak area: target selectivity and concentration-dependent enrichment. We benchmark pyBinder utilizing AS-MS data developed against antihemagglutinin antibody 12ca5, revealing that peptides that contain a motif known for target-specific high-affinity binding are well characterized by these two scores. AS-MS data from a second protein target, WD Repeat Domain 5 (WDR5), is analyzed to confirm the two pyBinder scores reliably capture the target-specific motif-containing peptides. From the results delivered by pyBinder, a list of target-selective features is developed and fed back into subsequent MS experiments to facilitate expanded data generation and the targeted discovery of selective ligands. pyBinder analysis resulted in a 4-fold increase in motif-containing sequence identification for WDR5 (from 3 to 14 ligands discovered), showing the utility of the two scores. This work establishes an improved approach for AS-MS to enable discovery outcomes (i.e., more ligands identified), but also a way to compare AS-MS data across samples, protocols, and conditions broadly.
Collapse
Affiliation(s)
- Joseph S Brown
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Michael A Lee
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Wayne Vuong
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Andrei Loas
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Bradley L Pentelute
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
149
|
Tolve M, Tutas J, Özer-Yildiz E, Klein I, Petzold A, Fritz VJ, Overhoff M, Silverman Q, Koletsou E, Liebsch F, Schwarz G, Korotkova T, Valtcheva S, Gatto G, Kononenko NL. The endocytic adaptor AP-2 maintains Purkinje cell function by balancing cerebellar parallel and climbing fiber synapses. Cell Rep 2025; 44:115256. [PMID: 39918958 DOI: 10.1016/j.celrep.2025.115256] [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: 06/11/2024] [Revised: 12/02/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
The loss of cerebellar Purkinje cells is a hallmark of neurodegenerative movement disorders, but the mechanisms remain enigmatic. We show that endocytic adaptor protein complex 2 (AP-2) is crucial for Purkinje cell survival. Using mouse genetics, viral tracing, calcium imaging, and kinematic analysis, we demonstrate that loss of the AP-2 μ subunit in Purkinje cells leads to early-onset ataxia and progressive degeneration. Synaptic dysfunction, marked by an overrepresentation of parallel fibers (PFs) over climbing fibers (CFs), precedes Purkinje cell loss. Mechanistically, AP-2 interacts with the PF-enriched protein GRID2IP, and its loss triggers GRID2IP degradation and glutamate δ2 receptor (GLURδ2) accumulation, leading to an excess of PFs while CFs are reduced. The overrepresentation of PFs increases Purkinje cell network activity, which is mitigated by enhancing glutamate clearance with ceftriaxone. These findings highlight the role of AP-2 in regulating GRID2IP levels in Purkinje cells to maintain PF-CF synaptic balance and prevent motor dysfunction.
Collapse
Affiliation(s)
- Marianna Tolve
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Janine Tutas
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ebru Özer-Yildiz
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ines Klein
- Neurology Department, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Anne Petzold
- Institute for Systems Physiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Veronika J Fritz
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Melina Overhoff
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Quinn Silverman
- Neurology Department, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Ellie Koletsou
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Filip Liebsch
- Institute of Biochemistry, Department of Chemistry, University of Cologne, Cologne, Germany
| | - Guenter Schwarz
- Institute of Biochemistry, Department of Chemistry, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Tatiana Korotkova
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Institute for Systems Physiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Silvana Valtcheva
- Institute for Systems Physiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Graziana Gatto
- Neurology Department, University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Natalia L Kononenko
- Cologne Excellence Cluster Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany; Center for Physiology and Pathophysiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Institute of Genetics, Faculty of Mathematics and Natural Sciences, University of Cologne, Cologne, Germany.
| |
Collapse
|
150
|
Arnold L, Yap M, Farrokhian N, Jackson L, Barry M, Ly T, Arjunan P, Kaczorowski-Worthley A, Tews C, Pandey A, Morrison A, Washburn MP, Standing D, Gomez JP, Yellapu NK, Johnson D, Li L, Umar S, Anant S, Thomas SM. DCLK1-mediated regulation of invadopodia dynamics and matrix metalloproteinase trafficking drives invasive progression in head and neck squamous cell carcinoma. Mol Cancer 2025; 24:50. [PMID: 39994636 PMCID: PMC11853957 DOI: 10.1186/s12943-025-02264-3] [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: 04/12/2024] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND HNSCC presents a significant health challenge due to its high mortality resulting from treatment resistance and locoregional invasion into critical structures in the head and neck region. Understanding the invasion mechanisms of HNSCC has the potential to guide targeted therapies, improving patient survival. Previously, we demonstrated the involvement of doublecortin like kinase 1 (DCLK1) in regulating HNSCC cell invasion. Here, we investigated the hypothesis that DCLK1 modulates proteins within invadopodia, specialized subcellular protrusions that secrete matrix metalloproteinases to degrade the ECM. METHODS We employed tandem mass tag (TMT)-based proteomics to identify the role of DCLK1 in regulating proteins involved in HNSCC invasion and validated the findings using immunoblotting. The Cancer Genome Atlas (TCGA) database was interrogated to correlate DCLK1 expression with tumor stage, grade, and invasion-associated proteins. In vitro invasion was assessed using a Boyden chamber assay, and immunohistochemistry on patient samples determined DCLK1's distribution within tumors. Gelatin invadopodia assay was used to establish DCLK1 localization to invadopodia related gelatin degradation. Super-resolution confocal microscopy demonstrated colocalization of DCLK1 with invadopodia markers and MMP trafficking proteins. ECM degradation by MMPs in HNSCC cells with wild-type and knockdown DCLK1 was evaluated using a dye-quenched tracer, while gel zymography and MMP array identified secreted proteases. Proximity ligation assay (PLA) and co-immunoprecipitation assays were used to confirm interactions between DCLK1, MMP9, KIF16B, and RAB40B. RESULTS Proteomic analysis demonstrate DCLK1's role in regulating proteins involved in cytoskeletal and ECM remodeling. Clinically, rising DCLK1 levels correlate with higher histological grade and lymph node metastasis, with heightened expression observed at the leading edge of HNSCC patient tissue. DCLK1 is localized with markers of mature invadopodia including TKS4, TKS5, cortactin, and MT1-MMP. Knockdown of DCLK1 led to reductions in invadopodia numbers and decreased in vitro invasion and ECM degradation. MMP9 colocalizes with DCLK1 within invadopodia structures and its secretion is disrupted by DCLK1 knockdown. Further, PLA and co-immunoprecipitations studies demonstrate DLCK1 complexes with KIF16B and RAB40B enabling trafficking of degradative MMP9 cargo along the invadopodia to degrade local ECM. CONCLUSION This work unveils a novel function of DCLK1 in regulating KIF16B and RAB40B to traffic matrix degrading MMP9 cargo to the distal end of the invadopodia facilitating HNSCC invasion.
Collapse
Affiliation(s)
- Levi Arnold
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
- Department of Cancer Biology, Medical Center, University of Kansas, Kansas, KS, 66160, USA
| | - Marrion Yap
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
| | - Nathan Farrokhian
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
| | - Laura Jackson
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
| | - Michael Barry
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
| | - Thuc Ly
- Department of Cancer Biology, Medical Center, University of Kansas, Kansas, KS, 66160, USA
| | - Pachiappan Arjunan
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
| | - Angela Kaczorowski-Worthley
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
| | - Carter Tews
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
| | - Avisha Pandey
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
| | - Austin Morrison
- Department of Cancer Biology, Medical Center, University of Kansas, Kansas, KS, 66160, USA
| | - Michael P Washburn
- Department of Cancer Biology, Medical Center, University of Kansas, Kansas, KS, 66160, USA
| | - David Standing
- Department of Cancer Biology, Medical Center, University of Kansas, Kansas, KS, 66160, USA
| | - Juan P Gomez
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA
| | - Nanda Kumar Yellapu
- Department of Biostatistics and Data Science, Medical Center, University of Kansas, Kansas, KS, 66160, USA
| | - David Johnson
- Computational Chemical Biology, University of Kansas, Lawrence, KS, 66047, USA
| | | | - Shahid Umar
- Department of Surgery, Medical Center, University of Kansas, Kansas, KS, 66160, USA
| | - Shrikant Anant
- Department of Cancer Biology, Medical Center, University of Kansas, Kansas, KS, 66160, USA
| | - Sufi Mary Thomas
- Department of Otolaryngology, Rainbow Blvd, University of Kansas Medical Center, 3901aq, Wahl Hall East 4031, Kansas, KS, 66160, USA.
- Department of Cancer Biology, Medical Center, University of Kansas, Kansas, KS, 66160, USA.
| |
Collapse
|