151
|
Opuni KFM, Ruß M, Geens R, Vocht LD, Wielendaele PV, Debuy C, Sterckx YGJ, Glocker MO. Mass spectrometry-complemented molecular modeling predicts the interaction interface for a camelid single-domain antibody targeting the Plasmodium falciparum circumsporozoite protein's C-terminal domain. Comput Struct Biotechnol J 2024; 23:3300-3314. [PMID: 39296809 PMCID: PMC11409006 DOI: 10.1016/j.csbj.2024.08.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 09/21/2024] Open
Abstract
Background Bioanalytical methods that enable rapid and high-detail characterization of binding specificities and strengths of protein complexes with low sample consumption are highly desired. The interaction between a camelid single domain antibody (sdAbCSP1) and its target antigen (PfCSP-Cext) was selected as a model system to provide proof-of-principle for the here described methodology. Research design and methods The structure of the sdAbCSP1 - PfCSP-Cext complex was modeled using AlphaFold2. The recombinantly expressed proteins, sdAbCSP1, PfCSP-Cext, and the sdAbCSP1 - PfCSP-Cext complex, were subjected to limited proteolysis and mass spectrometric peptide analysis. ITEM MS (Intact Transition Epitope Mapping Mass Spectrometry) and ITC (Isothermal Titration Calorimetry) were applied to determine stoichiometry and binding strength. Results The paratope of sdAbCSP1 mainly consists of its CDR3 (aa100-118). PfCSP-Cext's epitope is assembled from its α-helix (aa40-52) and opposing loop (aa83-90). PfCSP-Cext's GluC cleavage sites E46 and E58 were shielded by complex formation, confirming the predicted epitope. Likewise, sdAbCSP1's tryptic cleavage sites R105 and R108 were shielded by complex formation, confirming the predicted paratope. ITEM MS determined the 1:1 stoichiometry and the high complex binding strength, exemplified by the gas phase dissociation reaction enthalpy of 50.2 kJ/mol. The in-solution complex dissociation constant is 5 × 10-10 M. Conclusions Combining AlphaFold2 modeling with mass spectrometry/limited proteolysis generated a trustworthy model for the sdAbCSP1 - PfCSP-Cext complex interaction interface.
Collapse
Affiliation(s)
- Kwabena F M Opuni
- Department of Pharmaceutical Chemistry, School of Pharmacy, College of Health Science, University of Ghana, P.O. Box LG43, Legon, Ghana
| | - Manuela Ruß
- Proteome Center Rostock, University Medicine Rostock and University of Rostock, Schillingallee 69, 18057 Rostock, Germany
| | - Rob Geens
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical, and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, 2610 Antwerp, Belgium
| | - Line De Vocht
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical, and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, 2610 Antwerp, Belgium
| | - Pieter Van Wielendaele
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical, and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, 2610 Antwerp, Belgium
| | - Christophe Debuy
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical, and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, 2610 Antwerp, Belgium
| | - Yann G-J Sterckx
- Laboratory of Medical Biochemistry, Faculty of Pharmaceutical, Biomedical, and Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Wilrijk, 2610 Antwerp, Belgium
| | - Michael O Glocker
- Proteome Center Rostock, University Medicine Rostock and University of Rostock, Schillingallee 69, 18057 Rostock, Germany
| |
Collapse
|
152
|
Mares RG, Suica VI, Uyy E, Boteanu RM, Ivan L, Cocuz IG, Sabau AH, Yadav V, Szabo IA, Cotoi OS, Tomut ME, Jakobsson G, Simionescu M, Antohe F, Schiopu A. Short-term S100A8/A9 Blockade Promotes Cardiac Neovascularization after Myocardial Infarction. J Cardiovasc Transl Res 2024; 17:1389-1399. [PMID: 39009944 PMCID: PMC11634919 DOI: 10.1007/s12265-024-10542-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024]
Abstract
Acute-phase inhibition of the pro-inflammatory alarmin S100A8/A9 improves cardiac function post-myocardial infarction (MI), but the mechanisms underlying the long-term benefits of this short-term treatment remain to be elucidated. Here, we assessed the effects of S100A8/A9 blockade with the small-molecule inhibitor ABR-238901 on myocardial neovascularization in mice with induced MI. The treatment significantly reduced S100A9 and increased neovascularization in the myocardium, assessed by CD31 staining. Proteomic analysis by mass-spectrometry showed strong myocardial upregulation of the pro-angiogenic proteins filamin A (~ 10-fold) and reticulon 4 (~ 5-fold), and downregulation of the anti-angiogenic proteins Ras homolog gene family member A (RhoA, ~ 4.7-fold), neutrophilic granule protein (Ngp, ~ 4.0-fold), and cathelicidin antimicrobial peptide (Camp, ~ 4.4-fold) versus controls. In-vitro, ABR-238901 protected against apoptosis induced by recombinant human S100A8/A9 in human umbilical vein endothelial cells (HUVECs). In conclusion, S100A8/A9 blockade promotes post-MI myocardial neovascularization by favorably modulating pro-angiogenic proteins in the myocardium and by inhibiting endothelial cell apoptosis.
Collapse
Affiliation(s)
- Razvan Gheorghita Mares
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania.
| | - Viorel Iulian Suica
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Elena Uyy
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Raluca Maria Boteanu
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Luminita Ivan
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Iuliu Gabriel Cocuz
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania
- Clinical County Hospital, Targu Mures, Romania
| | - Adrian Horatiu Sabau
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania
- Clinical County Hospital, Targu Mures, Romania
| | - Vikas Yadav
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Istvan Adorjan Szabo
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania
| | - Ovidiu Simion Cotoi
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania
- Clinical County Hospital, Targu Mures, Romania
| | | | - Gabriel Jakobsson
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Maya Simionescu
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Felicia Antohe
- Department of Proteomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Alexandru Schiopu
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, Targu Mures, Romania.
- Molecular and Cellular Pharmacology - Functional Genomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania.
- Department of Translational Medicine, Lund University, Malmö, Sweden.
- Department of Internal Medicine, Skane University Hospital, Lund, Sweden.
| |
Collapse
|
153
|
Zuniga NR, Earls NE, Denos AEA, Elison JM, Jones BS, Smith EG, Moran NG, Broce KL, Romero GM, Hyer CD, Wagstaff KB, Almughamsi HM, Transtrum MK, Price JC. Quantitative and Kinetic Proteomics Reveal ApoE Isoform-dependent Proteostasis Adaptations in Mouse Brain. PLoS Comput Biol 2024; 20:e1012407. [PMID: 39666759 DOI: 10.1371/journal.pcbi.1012407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/26/2024] [Accepted: 11/03/2024] [Indexed: 12/14/2024] Open
Abstract
Apolipoprotein E (ApoE) polymorphisms modify the risk of Alzheimer's disease with ApoE4 strongly increasing and ApoE2 modestly decreasing risk relative to the control ApoE3. To investigate how ApoE isoforms alter risk, we measured changes in proteome homeostasis in transgenic mice expressing a human ApoE gene (isoform 2, 3, or 4). The regulation of each protein's homeostasis is observed by measuring turnover rate and abundance for that protein. We identified 4849 proteins and tested for ApoE isoform-dependent changes in the homeostatic regulation of ~2700 ontologies. In the brain, we found that ApoE4 and ApoE2 both lead to modified regulation of mitochondrial membrane proteins relative to the wild-type control ApoE3. In ApoE4 mice, lack of cohesion between mitochondrial membrane and matrix proteins suggests that dysregulation of proteasome and autophagy is reducing protein quality. In ApoE2, proteins of the mitochondrial matrix and the membrane, including oxidative phosphorylation complexes, had a similar increase in degradation which suggests coordinated replacement of the entire organelle. In the liver we did not observe these changes suggesting that the ApoE-effect on proteostasis is amplified in the brain relative to other tissues. Our findings underscore the utility of combining protein abundance and turnover rates to decipher proteome regulatory mechanisms and their potential role in biology.
Collapse
Affiliation(s)
- Nathan R Zuniga
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Noah E Earls
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Ariel E A Denos
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Jared M Elison
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Benjamin S Jones
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Ethan G Smith
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Noah G Moran
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Katie L Broce
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Gerome M Romero
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Chad D Hyer
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Kimberly B Wagstaff
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - Haifa M Almughamsi
- Department of Chemistry, College of Science, Taif University, Taif, Saudi Arabia
| | - Mark K Transtrum
- Department of Physics and Astronomy, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| | - John C Price
- Department of Chemistry and Biochemistry, College of Computational, Physical, and Mathematical Sciences, Brigham Young University, Provo, Utah, United States of America
| |
Collapse
|
154
|
Lord S, Johnston H, Samant R, Lai Y. Ubiquitylomics: An Emerging Approach for Profiling Protein Ubiquitylation in Skeletal Muscle. J Cachexia Sarcopenia Muscle 2024; 15:2281-2294. [PMID: 39279720 PMCID: PMC11634490 DOI: 10.1002/jcsm.13601] [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: 02/08/2024] [Revised: 07/18/2024] [Accepted: 08/12/2024] [Indexed: 09/18/2024] Open
Abstract
Skeletal muscle is a highly adaptable tissue, finely tuned by various physiological and pathological factors. Whilst the pivotal role of skeletal muscle in overall health is widely acknowledged, unravelling the underlying molecular mechanisms poses ongoing challenges. Protein ubiquitylation, a crucial post-translational modification, is involved in regulating most biological processes. This widespread impact is achieved through a diverse set of enzymes capable of generating structurally and functionally distinct ubiquitin modifications on proteins. The complexity of protein ubiquitylation has presented significant challenges in not only identifying ubiquitylated proteins but also characterising their functional significance. Mass spectrometry enables in-depth analysis of proteins and their post-translational modification status, offering a powerful tool for studying protein ubiquitylation and its biological diversity: an approach termed ubiquitylomics. Ubiquitylomics has been employed to tackle different perspectives of ubiquitylation, including but not limited to global quantification of substrates and ubiquitin linkages, ubiquitin site recognition and crosstalk with other post-translational modifications. As the field of mass spectrometry continues to evolve, the usage of ubiquitylomics has unravelled novel insights into the regulatory mechanisms of protein ubiquitylation governing biology. However, ubiquitylomics research has predominantly been conducted in cellular models, limiting our understanding of ubiquitin signalling events driving skeletal muscle biology. By integrating the intricate landscape of protein ubiquitylation with dynamic shifts in muscle physiology, ubiquitylomics promises to not only deepen our understanding of skeletal muscle biology but also lay the foundation for developing transformative muscle-related therapeutics. This review aims to articulate how ubiquitylomics can be utilised by researchers to address different aspects of ubiquitylation signalling in skeletal muscle. We explore methods used in ubiquitylomics experiments, highlight relevant literature employing ubiquitylomics in the context of skeletal muscle and outline considerations for experimental design.
Collapse
Affiliation(s)
- Samuel O. Lord
- School of Sport, Exercise and Rehabilitation SciencesUniversity of BirminghamBirminghamUK
| | | | | | - Yu‐Chiang Lai
- School of Sport, Exercise and Rehabilitation SciencesUniversity of BirminghamBirminghamUK
- MRC Versus Arthritis Centre for Musculoskeletal Ageing ResearchUniversity of BirminghamBirminghamUK
- NIHR Birmingham Biomedical Research Centre Sarcopenia and MultimorbidityUniversity of BirminghamBirminghamUK
| |
Collapse
|
155
|
Hamza H, Ghosh M, Löffler MW, Rammensee HG, Planz O. Identification and relative abundance of naturally presented and cross-reactive influenza A virus MHC class I-restricted T cell epitopes. Emerg Microbes Infect 2024; 13:2306959. [PMID: 38240239 PMCID: PMC10854457 DOI: 10.1080/22221751.2024.2306959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/14/2024] [Indexed: 02/10/2024]
Abstract
Cytotoxic T lymphocytes are key for controlling viral infection. Unravelling CD8+ T cell-mediated immunity to distinct influenza virus strains and subtypes across prominent HLA types is relevant for combating seasonal infections and emerging new variants. Using an immunopeptidomics approach, naturally presented influenza A virus-derived ligands restricted to HLA-A*24:02, HLA-A*68:01, HLA-B*07:02, and HLA-B*51:01 molecules were identified. Functional characterization revealed multifunctional memory CD8+ T cell responses for nine out of sixteen peptides. Peptide presentation kinetics was optimal around 12 h post infection and presentation of immunodominant epitopes shortly after infection was not always persistent. Assessment of immunogenic epitopes revealed that they are highly conserved across the major zoonotic reservoirs and may contain a single substitution in the vicinity of the anchor residues. These findings demonstrate how the identified epitopes promote T cell pools, possibly cross-protective in individuals and can be potential targets for vaccination.
Collapse
Affiliation(s)
- Hazem Hamza
- Institute for Immunology, University of Tübingen, Tübingen, Germany
- Virology Laboratory, Environmental Research Division, National Research Centre, Giza, Egypt
| | - Michael Ghosh
- Institute for Immunology, University of Tübingen, Tübingen, Germany
| | - Markus W Löffler
- Institute for Immunology, University of Tübingen, Tübingen, Germany
- Institute for Clinical and Experimental Transfusion Medicine, Medical Faculty of Tübingen, Tübingen, Germany
- Centre for Clinical Transfusion Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Hans-Georg Rammensee
- Institute for Immunology, University of Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
- Cluster of Excellence CMFI (EXC2124) "Controlling Microbes to Fight Infections", University of Tübingen, Tübingen, Germany
| | - Oliver Planz
- Institute for Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
156
|
Gonçalves Dias M, Doss B, Rawat A, Siegel KR, Mahathanthrige T, Sklenar J, Rodriguez Gallo MC, Derbyshire P, Dharmasena T, Cameron E, Uhrig RG, Zipfel C, Menke FLH, Monaghan J. Subfamily C7 Raf-like kinases MRK1, RAF26, and RAF39 regulate immune homeostasis and stomatal opening in Arabidopsis thaliana. THE NEW PHYTOLOGIST 2024; 244:2278-2294. [PMID: 39449177 PMCID: PMC11579443 DOI: 10.1111/nph.20198] [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: 08/05/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024]
Abstract
The calcium-dependent protein kinase CPK28 regulates several stress pathways in multiple plant species. Here, we aimed to discover CPK28-associated proteins in Arabidopsis thaliana. We used affinity-based proteomics and identified several potential CPK28 binding partners, including the C7 Raf-like kinases MRK1, RAF26, and RAF39. We used biochemistry, genetics, and physiological assays to gain insight into their function. We define redundant roles for these kinases in stomatal opening, immune-triggered reactive oxygen species (ROS) production, and resistance to a bacterial pathogen. We report that CPK28 associates with and trans-phosphorylates RAF26 and RAF39, and that MRK1, RAF26, and RAF39 are active kinases that localize to endomembranes. Although Raf-like kinases share some features with mitogen-activated protein kinase kinase kinases (MKKKs), we found that MRK1, RAF26, and RAF39 are unable to trans-phosphorylate any of the 10 Arabidopsis mitogen-activated protein kinase kinases (MKKs). Overall, our study suggests that C7 Raf-like kinases associate with and are phosphorylated by CPK28, function redundantly in stomatal opening and immunity, and possess substrate specificities distinct from canonical MKKKs.
Collapse
Affiliation(s)
| | - Bassem Doss
- Department of BiologyQueen's UniversityKingstonONK7L 3N6Canada
| | - Anamika Rawat
- Department of BiologyQueen's UniversityKingstonONK7L 3N6Canada
| | | | | | - Jan Sklenar
- The Sainsbury LaboratoryUniversity of East Anglia, Norwich Research ParkNorwichNR4 7UHUK
| | | | - Paul Derbyshire
- The Sainsbury LaboratoryUniversity of East Anglia, Norwich Research ParkNorwichNR4 7UHUK
| | | | - Emma Cameron
- Department of BiologyQueen's UniversityKingstonONK7L 3N6Canada
| | - R. Glen Uhrig
- Department of Biological SciencesUniversity of AlbertaEdmontonABT6G 2E9Canada
| | - Cyril Zipfel
- The Sainsbury LaboratoryUniversity of East Anglia, Norwich Research ParkNorwichNR4 7UHUK
- Institute of Plant and Microbial Biology and Zurich‐Basel Plant Science CenterUniversity of ZurichZurich8008Switzerland
| | - Frank L. H. Menke
- The Sainsbury LaboratoryUniversity of East Anglia, Norwich Research ParkNorwichNR4 7UHUK
| | - Jacqueline Monaghan
- Department of BiologyQueen's UniversityKingstonONK7L 3N6Canada
- The Sainsbury LaboratoryUniversity of East Anglia, Norwich Research ParkNorwichNR4 7UHUK
| |
Collapse
|
157
|
Torres-Romero I, Légeret B, Bertrand M, Sorigue D, Damm A, Cuiné S, Veillet F, Blot C, Brugière S, Couté Y, Garneau MG, Kotapati HK, Xin Y, Xu J, Bates PD, Thiam AR, Beisson F, Li-Beisson Y. α/β hydrolase domain-containing protein 1 acts as a lysolipid lipase and is involved in lipid droplet formation. Natl Sci Rev 2024; 11:nwae398. [PMID: 39791125 PMCID: PMC11711679 DOI: 10.1093/nsr/nwae398] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 01/12/2025] Open
Abstract
Lipid droplets (LDs) are the major sites of lipid and energy homeostasis. However, few LD biogenesis proteins have been identified. Using model microalga Chlamydomonas, we show that ABHD1, an α/β-hydrolase domain-containing protein, is localized to the LD surface and stimulates LD formation through two actions: one enzymatic and one structural. The knockout mutants contained similar amounts of triacylglycerols (TAG) but their LDs showed a higher content of lyso-derivatives of betaine lipid diacylglyceryl-N,N,N-trimethylhomoserine (DGTS). Over-expression of ABHD1 increased LD abundance and boosted TAG content. Purified recombinant ABHD1 hydrolyzed lyso-DGTS, producing a free fatty acid and a glyceryltrimethylhomoserine. In vitro droplet-embedded vesicles showed that ABHD1 promoted LD emergence. Taken together, these results identify ABHD1 as a new player in LD formation by its lipase activity on lyso-DGTS and by its distinct biophysical property. This study further suggests that lipases targeted to LDs and able to act on their polar lipid coat may be interesting tools to promote LD assembly in eukaryotic cells.
Collapse
Affiliation(s)
- Ismael Torres-Romero
- Aix Marseille Univ, CEA, CNRS, Institute of Bioscience and Biotechnology of Aix Marseille, BIAM, Saint-Paul-Lez-Durance 13108, France
| | - Bertrand Légeret
- Aix Marseille Univ, CEA, CNRS, Institute of Bioscience and Biotechnology of Aix Marseille, BIAM, Saint-Paul-Lez-Durance 13108, France
| | - Marie Bertrand
- Aix Marseille Univ, CEA, CNRS, Institute of Bioscience and Biotechnology of Aix Marseille, BIAM, Saint-Paul-Lez-Durance 13108, France
| | - Damien Sorigue
- Aix Marseille Univ, CEA, CNRS, Institute of Bioscience and Biotechnology of Aix Marseille, BIAM, Saint-Paul-Lez-Durance 13108, France
| | - Alicia Damm
- Laboratoire de Physique de l'École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université de Paris Cité, Paris 75005, France
| | - Stéphan Cuiné
- Aix Marseille Univ, CEA, CNRS, Institute of Bioscience and Biotechnology of Aix Marseille, BIAM, Saint-Paul-Lez-Durance 13108, France
| | - Florian Veillet
- Aix Marseille Univ, CEA, CNRS, Institute of Bioscience and Biotechnology of Aix Marseille, BIAM, Saint-Paul-Lez-Durance 13108, France
| | - Carla Blot
- Aix Marseille Univ, CEA, CNRS, Institute of Bioscience and Biotechnology of Aix Marseille, BIAM, Saint-Paul-Lez-Durance 13108, France
| | - Sabine Brugière
- Univ. Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, FR2048, Grenoble 38000, France
| | - Yohann Couté
- Univ. Grenoble Alpes, INSERM, CEA, UMR BioSanté U1292, CNRS, CEA, FR2048, Grenoble 38000, France
| | - Matthew G Garneau
- Institute of Biological Chemistry, Washington State University, Pullman 99164, USA
| | - Hari K Kotapati
- Institute of Biological Chemistry, Washington State University, Pullman 99164, USA
| | - Yi Xin
- Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
| | - Jian Xu
- Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
| | - Philip D Bates
- Institute of Biological Chemistry, Washington State University, Pullman 99164, USA
| | - Abdou R Thiam
- Laboratoire de Physique de l'École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université de Paris Cité, Paris 75005, France
| | - Fred Beisson
- Aix Marseille Univ, CEA, CNRS, Institute of Bioscience and Biotechnology of Aix Marseille, BIAM, Saint-Paul-Lez-Durance 13108, France
| | - Yonghua Li-Beisson
- Aix Marseille Univ, CEA, CNRS, Institute of Bioscience and Biotechnology of Aix Marseille, BIAM, Saint-Paul-Lez-Durance 13108, France
| |
Collapse
|
158
|
Svecla M, Moregola A, Dalt LD, Nour J, Baragetti A, Uboldi P, Idini A, Wuhrer M, Beretta G, Falck D, Bonacina F, Norata GD. ASGR1 deficiency improves atherosclerosis but alters liver metabolism in ApoE -/- mice. Cardiovasc Diabetol 2024; 23:428. [PMID: 39616371 PMCID: PMC11608471 DOI: 10.1186/s12933-024-02507-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/07/2024] [Indexed: 01/30/2025] Open
Abstract
The asialoglycoprotein receptor 1 (ASGR1), a multivalent carbohydrate-binding receptor that primarily is responsible for recognizing and eliminating circulating glycoproteins with exposed galactose (Gal) or N-acetylgalactosamine (GalNAc) as terminal glycan residues, has been implicated in modulating the lipid metabolism and reducing cardiovascular disease burden. In this study, we investigated the impact of ASGR1 deficiency (ASGR1-/-) on atherosclerosis by evaluating its effects on plaque formation, lipid metabolism, circulating immunoinflammatory response, and circulating N-glycome under the hypercholesterolemic condition in ApoE-deficient mice. After 16 weeks of a western-type diet, ApoE-/-/ASGR1-/- mice presented lower plasma cholesterol and triglyceride levels compared to ApoE-/-. This was associated with reduced atherosclerotic plaque area and necrotic core formation. Interestingly, ApoE-/-/ASGR1-/- mice showed increased levels of circulating immune cells, increased AST/ALT ratio, and no changes in the N-glycome profile and liver morphology. The liver of ApoE-/-/ASGR1-/- mice, however, presented alterations in the metabolism of lipids, xenobiotics, and bile secretion, indicating broader alterations in liver homeostasis beyond lipids. These data suggest that improvements in circulating lipid metabolism and atherosclerosis in ASGR1 deficiency is paralleled by a deterioration of liver injury. These findings point to the need for additional evaluation before considering ASGR1 as a pharmacological target for dyslipidemia and cardiovascular disorders.
Collapse
Affiliation(s)
- Monika Svecla
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Annalisa Moregola
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Da Dalt
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Jasmine Nour
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Patrizia Uboldi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Alessandra Idini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Giangiacomo Beretta
- Department of Environmental Science and Policy, Università degli Studi di Milano, Milan, Italy
| | - David Falck
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
159
|
Heitmann JS, Jung S, Wacker M, Maringer Y, Nelde A, Bauer J, Denk M, Hoenisch-Gravel N, Richter M, Oezbek MT, Dubbelaar ML, Bilich T, Pumptow M, Martus P, Illerhaus G, Denzlinger C, Steinbach F, Aulitzky WE, Müller MR, Dörfel D, Rammensee H, Salih HR, Walz JS. Warehouse-based, immunopeptidome-guided design of personalised peptide vaccines shows feasibility in clinical trial evaluation in CLL patients. Front Immunol 2024; 15:1482715. [PMID: 39660140 PMCID: PMC11628388 DOI: 10.3389/fimmu.2024.1482715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 10/30/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer peptide vaccination represents a promising therapeutic approach, but has been hampered by lack of suitable antigens and restricted applicability due to different HLA backgrounds of individual patients. We here introduce a novel warehouse-based concept for composition of personalized peptide vaccines and report on its successful application in a Phase II clinical trial in patients with chronic lymphocytic leukemia (CLL) after first-line therapy. 26 CLL patients in at least partial remission (PR) after 6 months of immuno-chemotherapy were vaccinated with a personalized vaccine compiled from a premanufactured peptide warehouse comprising immunopeptidome-defined CLL-associated peptides. Primary objective was evaluation of immunogenicity, secondary objectives were safety and minimal residual disease (MRD) response. Immunopeptidome-guided vaccine composition was throughout successful, proving the feasibility of warehouse-based vaccine design. Vaccination was well tolerated, with local injection site reactions being the most common adverse event. Only few patients showed vaccine-induced T cell responses, attributable to their inability to mount strong immune responses due to immune-chemotherapy and lack of potent adjuvant formulations. Both issues are addressed within a follow-up trial (NCT04688385), combining the immunopeptidome-guided warehouse-based vaccine design reported here with a potent novel adjuvant evaluating personalized multi- peptide vaccination in CLL patients under T cell supportive BTK inhibitor therapies. Clinical trial registration www.clinicaltrialsregister.eu, identifier NCT02802943.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Cancer Vaccines/immunology
- Cancer Vaccines/adverse effects
- Precision Medicine
- Vaccines, Subunit/immunology
- Aged
- Male
- Female
- Middle Aged
- Feasibility Studies
- Neoplasm, Residual
- Antigens, Neoplasm/immunology
- Aged, 80 and over
- Adult
- Protein Subunit Vaccines
Collapse
Affiliation(s)
- Jonas S. Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
| | - Susanne Jung
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
| | - Marcel Wacker
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
| | - Yacine Maringer
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
| | - Annika Nelde
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
| | - Jens Bauer
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Monika Denk
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Naomi Hoenisch-Gravel
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
| | - Marion Richter
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Melek T. Oezbek
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
| | - Marissa L. Dubbelaar
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
- Quantitative Biology Center (QBiC), University of Tübingen, Tübingen, Germany
| | - Tatjana Bilich
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Marina Pumptow
- Institute for Clinical Epidemiology and Applied Biometry, University Hospital Tübingen, Tübingen, Germany
| | - Peter Martus
- Institute for Clinical Epidemiology and Applied Biometry, University Hospital Tübingen, Tübingen, Germany
| | - Gerald Illerhaus
- Department of Hematology, Oncology and Palliative Care, Katharinenhospital Stuttgart, Stuttgart, Germany
| | - Claudio Denzlinger
- Department of Oncology, Hematology and Palliative Care, Marienhospital Stuttgart, Stuttgart, Germany
| | - Francesca Steinbach
- Department of Hematology, Oncology and Palliative Care, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Walter-Erich Aulitzky
- Department of Hematology, Oncology and Palliative Care, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | - Martin R. Müller
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Daniela Dörfel
- Department of Internal Medicine II, Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Hans–Georg Rammensee
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
- Institute of Immunology, University of Tübingen, Tübingen, Germany
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| | - Juliane S. Walz
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| |
Collapse
|
160
|
Olaisen C, Røst LM, Sharma A, Søgaard CK, Khong T, Berg S, Jang M, Nedal A, Spencer A, Bruheim P, Otterlei M. Multiple Myeloma Cells with Increased Proteasomal and ER Stress Are Hypersensitive to ATX-101, an Experimental Peptide Drug Targeting PCNA. Cancers (Basel) 2024; 16:3963. [PMID: 39682151 DOI: 10.3390/cancers16233963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Objectives: To examine the regulatory role of PCNA in MM, we have targeted PCNA with the experimental drug ATX-101 in three commercial cell lines (JJN3, RPMI 1660, AMO) and seven in-house patient-derived cell lines with a more primary cell-like phenotype (TK9, 10, 12, 13, 14, 16 and 18) and measured the systemic molecular effects. Methods: We have used a multi-omics untargeted approach, measuring the gene expression (transcriptomics), a subproteomics approach measuring mainly signalling proteins and proteins in complex with these (signallomics) and quantitative metabolomics. These results are supplemented with traditional analysis, e.g., viability, Western and ELISA analysis. Results: The sensitivity of the cell lines to ATX-101 varied, including between three cell lines derived from the same patient at different times of disease. A trend towards increased sensitivity to ATX-101 during disease progression was detected. Although with different sensitivities, ATX-101 treatment resulted in numerous changes in signalling and metabolite pools in all cell lines. Transcriptomics and signallomics analysis of the TK cell lines revealed that elevated endogenous expression of ribosomal genes, elevated proteasomal and endoplasmic reticulum (ER) stress and low endogenous levels of NAD+ and NADH were associated with ATX-101 hypersensitivity. ATX-101 treatment further enhanced the ER stress, reduced primary metabolism and reduced the levels of the redox pair GSH/GSSG in sensitive cells. Signallome analysis suggested that eleven proteins (TPD52, TNFRS17/BCMA, LILRB4/ILT3, TSG101, ZNRF2, UPF3B, FADS2, C11orf38/SMAP, CGREF1, GAA, COG4) were activated only in the sensitive MM cell lines (TK13, 14 and 16 and JJN3), and not in nine other cancer cell lines or in primary monocytes. These proteins may therefore be biomarkers of cells with activated proteasomal and ER stress even though the gene expression levels of these proteins were not elevated. Interestingly, carfilzomib-resistant cells were at least as sensitive to ATX-101 as the wild-type cells, suggesting both low cross-resistance between ATX-101 and proteasome inhibitors and elevated proteasomal stress in carfilzomib-resistant cells. Conclusions: Our multi-omics approach revealed a vital role of PCNA in regulation of proteasomal and ER stress in MM.
Collapse
Affiliation(s)
- Camilla Olaisen
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Lisa Marie Røst
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Animesh Sharma
- Proteomics and Modomics Experimental Core Facility (PROMEC), NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Caroline Krogh Søgaard
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Tiffany Khong
- Australian Centre for Blood Diseases, Monash University, Melbourne 3004, Australia
- Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne 3004, Australia
| | - Sigrid Berg
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Mi Jang
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Aina Nedal
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Andrew Spencer
- Australian Centre for Blood Diseases, Monash University, Melbourne 3004, Australia
- Department of Malignant Haematology and Stem Cell Transplantation, Alfred Hospital, Melbourne 3004, Australia
| | - Per Bruheim
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| | - Marit Otterlei
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
- Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, NO-7006 Trondheim, Norway
- APIM Therapeutics A/S, Rådhusveien 12, NO-7100 Rissa, Norway
| |
Collapse
|
161
|
Greiner-Tollersrud OK, Krausz M, Boehler V, Polyzou A, Seidl M, Spahiu A, Abdullah Z, Andryka-Cegielski K, Dominick FI, Huebscher K, Goschin A, Smulski CR, Trompouki E, Link R, Ebersbach H, Srinivas H, Marchant M, Sogkas G, Staab D, Vågbø C, Guerini D, Baasch S, Latz E, Hartmann G, Henneke P, Geiger R, Peng XP, Grimbacher B, Bartok E, Alseth I, Warncke M, Proietti M. ADA2 is a lysosomal deoxyadenosine deaminase acting on DNA involved in regulating TLR9-mediated immune sensing of DNA. Cell Rep 2024; 43:114899. [PMID: 39441717 DOI: 10.1016/j.celrep.2024.114899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 09/19/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
Although adenosine deaminase 2 (ADA2) is considered an extracellular ADA, evidence questions the physiological relevance of this activity. Our study reveals that ADA2 localizes within the lysosomes, where it is targeted through modifications of its glycan structures. We show that ADA2 interacts with DNA molecules, altering their sequences by converting deoxyadenosine (dA) to deoxyinosine (dI). We characterize its DNA substrate preferences and provide data suggesting that DNA, rather than free adenosine, is its natural substrate. Finally, we demonstrate that dA-to-dI editing of DNA molecules and ADA2 regulate lysosomal immune sensing of nucleic acids (NAs) by modulating Toll-like receptor 9 (TLR9) activation. Our results describe a mechanism involved in the complex interplay between NA metabolism and immune response, possibly impacting ADA2 deficiency (DADA2) and other diseases involving this pathway, including autoimmune diseases, cancer, or infectious diseases.
Collapse
Affiliation(s)
| | - Máté Krausz
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Vincent Boehler
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Aikaterini Polyzou
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; IRCAN Institute for Research on Cancer and Aging, INSERM Unité 1081, CNRS UMR 7284, Université Côte d'Azur, Nice, France
| | - Maximilian Seidl
- Institute of Pathology, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Ambra Spahiu
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Zeinab Abdullah
- Institute of Experimental Immunology, Universitätsklinikum Bonn, Bonn, Germany
| | | | | | - Katrin Huebscher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany; Institut für Forstentomologie und Waldschutz, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Andreas Goschin
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Cristian R Smulski
- Medical Physics Department, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Carlos de Bariloche, Río Negro, Argentina
| | - Eirini Trompouki
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany; IRCAN Institute for Research on Cancer and Aging, INSERM Unité 1081, CNRS UMR 7284, Université Côte d'Azur, Nice, France
| | - Regina Link
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Hilmar Ebersbach
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Honnappa Srinivas
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Martine Marchant
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Georgios Sogkas
- Department of Rheumatology and Clinical Immunology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; RESIST - Cluster of Excellence 2155, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Dieter Staab
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Cathrine Vågbø
- Proteomics and Modomics Experimental Core (PROMEC), Norwegian University of Science and Technology and the Central Norway Regional Health Authority, Trondheim, Norway
| | - Danilo Guerini
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Sebastian Baasch
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany; Institute for Infection Prevention and Control, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Freiburg Institute for Advanced Studies (FRIAS), University of Freiburg, Freiburg, Germany
| | - Eicke Latz
- Institute of Innate Immunity, Universitätsklinikum Bonn, Bonn, Germany
| | - Gunther Hartmann
- Institute of Experimental Hematology and Transfusion Medicine Bonn, Bonn, Germany
| | - Philippe Henneke
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany; Institute for Infection Prevention and Control, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Roger Geiger
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland; Institute of Oncology Research, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Xiao P Peng
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany; RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Freiburg, Germany
| | - Eva Bartok
- Institute of Experimental Hematology and Transfusion Medicine Bonn, Bonn, Germany
| | - Ingrun Alseth
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Max Warncke
- Novartis Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Michele Proietti
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany; Department of Rheumatology and Clinical Immunology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany; RESIST - Cluster of Excellence 2155, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
162
|
Takemon Y, Pleasance ED, Gagliardi A, Hughes CS, Csizmok V, Wee K, Trinh DL, Huff RD, Mungall AJ, Moore RA, Chuah E, Mungall KL, Lewis E, Nelson J, Lim HJ, Renouf DJ, Jones SJ, Laskin J, Marra MA. Mapping in silico genetic networks of the KMT2D tumour suppressor gene to uncover novel functional associations and cancer cell vulnerabilities. Genome Med 2024; 16:136. [PMID: 39578878 PMCID: PMC11583415 DOI: 10.1186/s13073-024-01401-9] [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: 08/05/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Loss-of-function (LOF) alterations in tumour suppressor genes cannot be directly targeted. Approaches characterising gene function and vulnerabilities conferred by such mutations are required. METHODS Here, we computationally map genetic networks of KMT2D, a tumour suppressor gene frequently mutated in several cancer types. Using KMT2D loss-of-function (KMT2DLOF) mutations as a model, we illustrate the utility of in silico genetic networks in uncovering novel functional associations and vulnerabilities in cancer cells with LOF alterations affecting tumour suppressor genes. RESULTS We revealed genetic interactors with functions in histone modification, metabolism, and immune response and synthetic lethal (SL) candidates, including some encoding existing therapeutic targets. Notably, we predicted WRN as a novel SL interactor and, using recently available WRN inhibitor (HRO761 and VVD-133214) treatment response data, we observed that KMT2D mutational status significantly distinguishes treatment-sensitive MSI cell lines from treatment-insensitive MSI cell lines. CONCLUSIONS Our study thus illustrates how tumour suppressor gene LOF alterations can be exploited to reveal potentially targetable cancer cell vulnerabilities.
Collapse
Affiliation(s)
- Yuka Takemon
- Genome Science and Technology Graduate Program, University of British Columbia, Vancouver, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Erin D Pleasance
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Alessia Gagliardi
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | | | - Veronika Csizmok
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Kathleen Wee
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Diane L Trinh
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Ryan D Huff
- Division of Respiratory Medicine, Department of Medicine, Air Pollution Exposure Laboratory, Vancouver Coastal Health Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Eric Chuah
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Eleanor Lewis
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Jessica Nelson
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
| | - Howard J Lim
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Daniel J Renouf
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
- Pancreas Centre BC, Vancouver, BC, Canada
| | - Steven Jm Jones
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Janessa Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Marco A Marra
- Michael Smith Laboratories, University of British Columbia, Vancouver, Canada.
- Canada's Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
163
|
Jørgensen SH, Emdal KB, Pedersen AK, Axelsen LN, Kildegaard HF, Demozay D, Pedersen TÅ, Grønborg M, Slaaby R, Nielsen PK, Olsen JV. Multi-layered proteomics identifies insulin-induced upregulation of the EphA2 receptor via the ERK pathway which is dependent on low IGF1R level. Sci Rep 2024; 14:28856. [PMID: 39572596 PMCID: PMC11582730 DOI: 10.1038/s41598-024-77817-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/25/2024] [Indexed: 11/24/2024] Open
Abstract
Insulin resistance impairs the cellular insulin response, and often precedes metabolic disorders, like type 2 diabetes, impacting an increasing number of people globally. Understanding the molecular mechanisms in hepatic insulin resistance is essential for early preventive treatments. To elucidate changes in insulin signal transduction associated with hepatocellular resistance, we employed a multi-layered mass spectrometry-based proteomics approach focused on insulin receptor (IR) signaling at the interactome, phosphoproteome, and proteome levels in a long-term hyperinsulinemia-induced insulin-resistant HepG2 cell line with a knockout of the insulin-like growth factor 1 receptor (IGF1R KO). The analysis revealed insulin-stimulated recruitment of the PI3K complex in both insulin-sensitive and -resistant cells. Phosphoproteomics showed attenuated signaling via the metabolic PI3K-AKT pathway but sustained extracellular signal-regulated kinase (ERK) activity in insulin-resistant cells. At the proteome level, the ephrin type-A receptor 2 (EphA2) showed an insulin-induced increase in expression, which occurred through the ERK signaling pathway and was concordantly independent of insulin resistance. Induction of EphA2 by insulin was confirmed in additional cell lines and observed uniquely in cells with high IR-to-IGF1R ratio. The multi-layered proteomics dataset provided insights into insulin signaling, serving as a resource to generate and test hypotheses, leading to an improved understanding of insulin resistance.
Collapse
Affiliation(s)
- Sarah Hyllekvist Jørgensen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
- Global Research Technologies, Novo Nordisk A/S, 2760, Maaloev, Denmark
| | - Kristina Bennet Emdal
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| | - Anna-Kathrine Pedersen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | | | | | - Damien Demozay
- Global Drug Discovery, Novo Nordisk A/S, 2760, Maaloev, Denmark
| | | | - Mads Grønborg
- Global Translation, Novo Nordisk A/S, 2760, Maaloev, Denmark
| | - Rita Slaaby
- Global Drug Discovery, Novo Nordisk A/S, 2760, Maaloev, Denmark
| | | | - Jesper Velgaard Olsen
- Proteomics Program, Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.
| |
Collapse
|
164
|
Kumar K, Pazare M, Ratnaparkhi GS, Kamat SS. CG17192 is a Phospholipase That Regulates Signaling Lipids in the Drosophila Gut upon Infection. Biochemistry 2024; 63:3000-3010. [PMID: 39442931 DOI: 10.1021/acs.biochem.4c00579] [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: 10/25/2024]
Abstract
The chemoproteomics technique, activity-based protein profiling (ABPP), has proven to be an invaluable tool in assigning functions to enzymes. The serine hydrolase (SH) enzyme superfamily, in particular, has served as an excellent example in displaying the versatility of various ABPP platforms and has resulted in a comprehensive cataloging of the biochemical activities associated within this superfamily. Besides SHs, in mammals, several other enzyme classes have been thoroughly investigated using ABPP platforms. However, the utility of ABPP platforms in fly models remains underexplored. Realizing this knowledge gap, leveraging complementary ABPP platforms, we reported the full array of SH activities during various developmental stages and adult tissues in the fruit fly (Drosophila melanogaster). Following up on this study, using ABPP, we mapped SH activities in adult fruit flies in an infection model and found that a gut-resident lipase CG17192 showed increased activity during infection. To assign a biological function to this uncharacterized lipase, we performed an untargeted lipidomics analysis and found that phosphatidylinositols were significantly elevated when CG17192 was depleted in the adult fruit fly gut. Next, we overexpressed this lipase in insect cells, and using biochemical assays, we show that CG17192 is a secreted enzyme that has phospholipase C (PLC) type activity, with phosphatidylinositol being a preferred substrate. Finally, we show during infection that heightened CG17192 regulates phosphatidylinositol levels and, by doing so, likely modulates signaling pathways in the adult fruit fly gut that might be involved in the resolution of this pathophysiological condition.
Collapse
Affiliation(s)
- Kundan Kumar
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Mrunal Pazare
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Girish S Ratnaparkhi
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| | - Siddhesh S Kamat
- Department of Biology, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pashan, Pune 411008, Maharashtra, India
| |
Collapse
|
165
|
Foged MM, Recazens E, Chollet S, Lisci M, Allen GE, Zinshteyn B, Boutguetait D, Münch C, Mootha VK, Jourdain AA. Cytosolic N6AMT1-dependent translation supports mitochondrial RNA processing. Proc Natl Acad Sci U S A 2024; 121:e2414187121. [PMID: 39503847 PMCID: PMC11588129 DOI: 10.1073/pnas.2414187121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/03/2024] [Indexed: 11/27/2024] Open
Abstract
Mitochondrial biogenesis relies on both the nuclear and mitochondrial genomes, and imbalance in their expression can lead to inborn errors of metabolism, inflammation, and aging. Here, we investigate N6AMT1, a nucleo-cytosolic methyltransferase that exhibits genetic codependency with mitochondria. We determine transcriptional and translational profiles of N6AMT1 and report that it is required for the cytosolic translation of TRMT10C (MRPP1) and PRORP (MRPP3), two subunits of the mitochondrial RNAse P enzyme. In the absence of N6AMT1, or when its catalytic activity is abolished, RNA processing within mitochondria is impaired, leading to the accumulation of unprocessed and double-stranded RNA, thus preventing mitochondrial protein synthesis and oxidative phosphorylation, and leading to an immune response. Our work sheds light on the function of N6AMT1 in protein synthesis and highlights a cytosolic program required for proper mitochondrial biogenesis.
Collapse
Affiliation(s)
- Mads M. Foged
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Emeline Recazens
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Sylvain Chollet
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Miriam Lisci
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - George E. Allen
- Department of Microbiology and Molecular Medicine, Institute of Genetics and Genomics Geneva, Faculty of Medicine, University of Geneva, Geneva 41211, Switzerland
| | - Boris Zinshteyn
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Doha Boutguetait
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main60590, Germany
| | - Christian Münch
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main60590, Germany
| | - Vamsi K. Mootha
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA02142
- HHMI, Massachusetts General Hospital Boston, MA02114
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA02114
- Department of Systems Biology, Harvard Medical School, Boston, MA02115
| | - Alexis A. Jourdain
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| |
Collapse
|
166
|
Kroh A, Schmitz S, Köhne S, Andruszkow J, Nolting J, Preisinger C, Große K, Eickhoff RM, Heise D, Cramer T, Rheinwalt KP, Alizai PH, Neumann UP, Ulmer TF. Sleeve-gastrectomy results in improved metabolism and a massive stress response of the liver proteome in a mouse model of metabolic dysfunction-associated steatohepatitis. Heliyon 2024; 10:e38678. [PMID: 39524892 PMCID: PMC11550656 DOI: 10.1016/j.heliyon.2024.e38678] [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: 03/12/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
Background Bariatric surgery has been shown to improve the histopathological findings in patients with obesity and metabolic dysfunction-associated steatohepatitis, but there are also reports about non-responders or progressive disease after bariatric interventions. Therefore, it is of utmost importance to understand the pathophysiological processes in the liver after bariatric surgery. Materials and methods In the present study, 4 weeks old male C57/Bl6 mice were fed a Western Diet to induce metabolic dysfunction-associated steatohepatitis and sleeve-gastrectomy (SG), or sham operation in the pair-fed and ad libitum control group were performed. Mice were observed for two or eight weeks after surgery and metabolic assessment was performed throughout the experiment. Histopathology, flow cytometry and proteomic analyses were conducted to evaluate hepatic inflammation, liver metabolism and affected signaling pathways. Results Weight loss was higher, and metabolism significantly improved after SG. Two weeks after SG major inflammatory and regulatory disturbances in the liver were observed. The proportion of hepatic CD3+NK1.1+ cells were decreased, and proteins involved in apoptosis like Fas, Casp1 and Casp9 or in the acute phase response were upregulated in SG mice. These disturbances decreased in the long-term and we observed an increase of many proteins involved in lipid metabolism eight weeks following SG. Conclusions The rapid weight loss and decrease of hepatic fat after SG lead to a proinflammatory response in the liver in the early phase after surgery, which changes to a more moderate immune response in the long-term. We suggest a preoperative risk stratification and postoperative surveillance depending on the histopathological findings.
Collapse
Affiliation(s)
- Andreas Kroh
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Sophia Schmitz
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
- General-, Visceral- and Transplant Surgery, Uniklinik Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Saskia Köhne
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Julia Andruszkow
- Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Jochen Nolting
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Christian Preisinger
- Proteomics Facility, Interdisciplinary Center for Clinical Research (IZKF) Aachen, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Karsten Große
- Department of Medicine III, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Roman M. Eickhoff
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Daniel Heise
- General-, Visceral- and Transplant Surgery, Uniklinik Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Thorsten Cramer
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Karl Peter Rheinwalt
- Department for Bariatric and Metabolic Surgery, St. Franziskus Hospital, Cologne, Germany
| | - Patrick Hamid Alizai
- Department of General and Visceral Surgery, Gemeinschaftskrankenhaus Bonn, Bonn, Germany
| | - Ulf Peter Neumann
- General-, Visceral- and Transplant Surgery, Uniklinik Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Tom Florian Ulmer
- General-, Visceral- and Transplant Surgery, Uniklinik Essen, Hufelandstr. 55, 45147, Essen, Germany
| |
Collapse
|
167
|
Stauberová V, Kubeša B, Joseph M, Benedet M, Furlan B, Buriánková K, Ulrych A, Kupčík R, Vomastek T, Massidda O, Tsui HCT, Winkler ME, Branny P, Doubravová L. GpsB Coordinates StkP Signaling as a PASTA Kinase Adaptor in Streptococcus pneumoniae Cell Division. J Mol Biol 2024; 436:168797. [PMID: 39303764 PMCID: PMC11563889 DOI: 10.1016/j.jmb.2024.168797] [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: 05/09/2024] [Revised: 09/05/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
StkP, the Ser/Thr protein kinase of the major human pathogen Streptococcus pneumoniae, monitors cell wall signals and regulates growth and division in response. In vivo, StkP interacts with GpsB, a cell division protein required for septal ring formation and closure, that affects StkP-dependent phosphorylation. Here, we report that although StkP has basal intrinsic kinase activity, GpsB promotes efficient autophosphorylation of StkP and phosphorylation of StkP substrates. Phosphoproteomic analyzes showed that GpsB is phosphorylated at several Ser and Thr residues. We confirmed that StkP directly phosphorylates GpsB in vitro and in vivo, with T79 and T83 being the major phosphorylation sites. In vitro, phosphoablative GpsB substitutions had a lower potential to stimulate StkP activity, whereas phosphomimetic substitutions were functional in terms of StkP activation. In vivo, substitutions of GpsB phosphoacceptor residues, either phosphoablative or mimetic, had a negative effect on GpsB function, resulting in reduced StkP-dependent phosphorylation and impaired cell division. The bacterial two-hybrid assay and co-immunoprecipitation of GpsB from cells with differentially active StkP indicated that increased phosphorylation of GpsB resulted in a more efficient interaction of GpsB with StkP. Our data suggest that GpsB acts as an adaptor that directly promotes StkP activity by mediating interactions within the StkP signaling hub, ensuring StkP recruitment into the complex and substrate specificity. We present a model that interaction of StkP with GpsB and its phosphorylation and dephosphorylation dynamically modulate kinase activity during exponential growth and under cell wall stress of S. pneumoniae, ensuring the proper functioning of the StkP signaling pathway.
Collapse
Affiliation(s)
- Václava Stauberová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Bohumil Kubeša
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Merrin Joseph
- Department of Biology, Indiana University Bloomington, 1001 E 3rd Street, Bloomington, IN 47405-7005, USA
| | - Mattia Benedet
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Berenice Furlan
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Karolína Buriánková
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Aleš Ulrych
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Rudolf Kupčík
- Biomedical Research Centre, University Hospital Hradec Králové, 500 05 Hradec Králové, Czech Republic
| | - Tomáš Vomastek
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Orietta Massidda
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy
| | - Ho-Ching T Tsui
- Department of Biology, Indiana University Bloomington, 1001 E 3rd Street, Bloomington, IN 47405-7005, USA
| | - Malcolm E Winkler
- Department of Biology, Indiana University Bloomington, 1001 E 3rd Street, Bloomington, IN 47405-7005, USA
| | - Pavel Branny
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Linda Doubravová
- Institute of Microbiology of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic.
| |
Collapse
|
168
|
Prieto-Garcia C, Matkovic V, Mosler T, Li C, Liang J, Oo JA, Haidle F, Mačinković I, Cabrera-Orefice A, Berkane R, Giuliani G, Xu F, Jacomin AC, Tomaskovic I, Basoglu M, Hoffmann ME, Rathore R, Cetin R, Boutguetait D, Bozkurt S, Hernández Cañás MC, Keller M, Busam J, Shah VJ, Wittig I, Kaulich M, Beli P, Galej WP, Ebersberger I, Wang L, Münch C, Stolz A, Brandes RP, Tse WKF, Eimer S, Stainier DYR, Legewie S, Zarnack K, Müller-McNicoll M, Dikic I. Pathogenic proteotoxicity of cryptic splicing is alleviated by ubiquitination and ER-phagy. Science 2024; 386:768-776. [PMID: 39541449 DOI: 10.1126/science.adi5295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/22/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
RNA splicing enables the functional adaptation of cells to changing contexts. Impaired splicing has been associated with diseases, including retinitis pigmentosa, but the underlying molecular mechanisms and cellular responses remain poorly understood. In this work, we report that deficiency of ubiquitin-specific protease 39 (USP39) in human cell lines, zebrafish larvae, and mice led to impaired spliceosome assembly and a cytotoxic splicing profile characterized by the use of cryptic 5' splice sites. Disruptive cryptic variants evaded messenger RNA (mRNA) surveillance pathways and were translated into misfolded proteins, which caused proteotoxic aggregates, endoplasmic reticulum (ER) stress, and, ultimately, cell death. The detrimental consequence of splicing-induced proteotoxicity could be mitigated by up-regulating the ubiquitin-proteasome system and selective autophagy. Our findings provide insight into the molecular pathogenesis of spliceosome-associated diseases.
Collapse
Affiliation(s)
- Cristian Prieto-Garcia
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Vigor Matkovic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Thorsten Mosler
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Congxin Li
- Department of Systems Biology and Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| | - Jie Liang
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - James A Oo
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany
- Cardiopulmonary Institute (CPI), Goethe University Frankfurt, Frankfurt, Germany
| | - Felix Haidle
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Igor Mačinković
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Alfredo Cabrera-Orefice
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt, Germany
- Center for Functional Proteomics, Goethe University Frankfurt, Frankfurt, Germany
| | - Rayene Berkane
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Giulio Giuliani
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Fenfen Xu
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P.R. China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Anne-Claire Jacomin
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Ines Tomaskovic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Marion Basoglu
- Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt, Germany
| | - Marina E Hoffmann
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Rajeshwari Rathore
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Ronay Cetin
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Doha Boutguetait
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Süleyman Bozkurt
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Mario Keller
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Jonas Busam
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Varun Jayeshkumar Shah
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Ilka Wittig
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany
- Center for Functional Proteomics, Goethe University Frankfurt, Frankfurt, Germany
| | - Manuel Kaulich
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Petra Beli
- Institute of Molecular Biology (IMB), Mainz, Germany
- Institute of Developmental Biology and Neurobiology (IDN), Johannes Gutenberg-University, Mainz, Germany
| | | | - Ingo Ebersberger
- Applied Bioinformatics Group, Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt, Germany
- Senckenberg Biodiversity and Climate Research Centre (S-BIK-F), Frankfurt, Germany
- LOEWE Centre for Translational Biodiversity Genomics (TBG), Frankfurt, Germany
| | - Likun Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P.R. China
| | - Christian Münch
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Center for Functional Proteomics, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Systems Medicine, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
| | - Alexandra Stolz
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe University Frankfurt, Frankfurt, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhine-Main, Frankfurt, Germany
- Cardiopulmonary Institute (CPI), Goethe University Frankfurt, Frankfurt, Germany
| | - William Ka Fai Tse
- Laboratory of Developmental Disorders and Toxicology, Center for Promotion of International Education and Research, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Stefan Eimer
- Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt, Germany
| | - Didier Y R Stainier
- Max Planck Institute for Heart and Lung Research, Department of Developmental Genetics, Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Bad Nauheim, Germany
- Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Stefan Legewie
- Department of Systems Biology and Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| | - Kathi Zarnack
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
| | - Michaela Müller-McNicoll
- Institute of Molecular Biosciences, Goethe University Frankfurt, Frankfurt, Germany
- Max-Planck Institute for Biophysics, Frankfurt, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt, Germany
- Max-Planck Institute for Biophysics, Frankfurt, Germany
| |
Collapse
|
169
|
Ashkarran AA, Gharibi H, Sadeghi SA, Modaresi SM, Wang Q, Lin TJ, Yerima G, Tamadon A, Sayadi M, Jafari M, Lin Z, Ritz D, Kakhniashvili D, Guha A, Mofrad MRK, Sun L, Landry MP, Saei AA, Mahmoudi M. Small molecule modulation of protein corona for deep plasma proteome profiling. Nat Commun 2024; 15:9638. [PMID: 39511193 PMCID: PMC11544298 DOI: 10.1038/s41467-024-53966-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 10/29/2024] [Indexed: 11/15/2024] Open
Abstract
The protein corona formed on nanoparticles (NPs) has potential as a valuable diagnostic tool for improving plasma proteome coverage. Here, we show that spiking small molecules, including metabolites, lipids, vitamins, and nutrients into plasma can induce diverse protein corona patterns on otherwise identical NPs, significantly enhancing the depth of plasma proteome profiling. The protein coronas on polystyrene NPs when exposed to plasma treated with an array of small molecules allows for the detection of 1793 proteins marking an 8.25-fold increase in the number of quantified proteins compared to plasma alone (218 proteins) and a 2.63-fold increase relative to the untreated protein corona (681 proteins). Furthermore, we discovered that adding 1000 µg/ml phosphatidylcholine could singularly enable the detection of 897 proteins. At this specific concentration, phosphatidylcholine selectively depletes the four most abundant plasma proteins, including albumin, thus reducing the dynamic range of plasma proteome and enabling the detection of proteins with lower abundance. Employing an optimized data-independent acquisition approach, the inclusion of phosphatidylcholine leads to the detection of 1436 proteins in a single plasma sample. Our molecular dynamics results reveal that phosphatidylcholine interacts with albumin via hydrophobic interactions, H-bonds, and water bridges. The addition of phosphatidylcholine also enables the detection of 337 additional proteoforms compared to untreated protein corona using a top-down proteomics approach. Given the critical role of plasma proteomics in biomarker discovery and disease monitoring, we anticipate the widespread adoption of this methodology for the identification and clinical translation of biomarkers.
Collapse
Affiliation(s)
- Ali Akbar Ashkarran
- Precision Health Program, Michigan State University, East Lansing, MI, USA
- Depatment of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Hassan Gharibi
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Qianyi Wang
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Teng-Jui Lin
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Ghafar Yerima
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Ali Tamadon
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Maryam Sayadi
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Maryam Jafari
- Division of ENT Diseases, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Zijin Lin
- Precision Health Program, Michigan State University, East Lansing, MI, USA
| | - Danilo Ritz
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - David Kakhniashvili
- Proteomics and Metabolomics Core Facility, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Avirup Guha
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, Berkeley, CA, USA
| | - Liangliang Sun
- Department of Chemistry, Michigan State University, East Lansing, MI, USA
| | - Markita P Landry
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
- Department of Neuroscience, University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Amir Ata Saei
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Morteza Mahmoudi
- Precision Health Program, Michigan State University, East Lansing, MI, USA.
- Depatment of Radiology, College of Human Medicine, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
170
|
Won SJ, Zhang Y, Reinhardt CJ, Hargis LM, MacRae NS, DeMeester KE, Njomen E, Remsberg JR, Melillo B, Cravatt BF, Erb MA. Redirecting the pioneering function of FOXA1 with covalent small molecules. Mol Cell 2024; 84:4125-4141.e10. [PMID: 39413792 PMCID: PMC11560529 DOI: 10.1016/j.molcel.2024.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/08/2024] [Accepted: 09/20/2024] [Indexed: 10/18/2024]
Abstract
Pioneer transcription factors (TFs) bind to and open closed chromatin, facilitating engagement by other regulatory factors involved in gene activation or repression. Chemical probes are lacking for pioneer TFs, which has hindered their mechanistic investigation in cells. Here, we report the chemical proteomic discovery of electrophilic compounds that stereoselectively and site-specifically bind the pioneer TF forkhead box protein A1 (FOXA1) at a cysteine (C258) within the forkhead DNA-binding domain. We show that these covalent ligands react with FOXA1 in a DNA-dependent manner and rapidly remodel its pioneer activity in prostate cancer cells reflected in redistribution of FOXA1 binding across the genome and directionally correlated changes in chromatin accessibility. Motif analysis supports a mechanism where the ligands relax the canonical DNA-binding preference of FOXA1 by strengthening interactions with suboptimal sequences in predicted proximity to C258. Our findings reveal a striking plasticity underpinning the pioneering function of FOXA1 that can be controlled by small molecules.
Collapse
Affiliation(s)
- Sang Joon Won
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yuxiang Zhang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | - Lauren M Hargis
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Nicole S MacRae
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Kristen E DeMeester
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Evert Njomen
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jarrett R Remsberg
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Bruno Melillo
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Michael A Erb
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
171
|
Rashmi U, Bhatia S, Nayak M, Khochare S, Sunagar K. Elusive elapids: biogeographic venom variation in Indian kraits and its repercussion on snakebite therapy. Front Pharmacol 2024; 15:1443073. [PMID: 39575383 PMCID: PMC11579489 DOI: 10.3389/fphar.2024.1443073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/21/2024] [Indexed: 11/24/2024] Open
Abstract
Snakebite is a major public health concern in many parts of the world, including India, where over 58,000 deaths occur annually due to snake envenoming. The common krait (Bungarus caeruleus) is responsible for the second-highest number of snakebite-related mortalities in the country. However, despite its notoriety, little is known about its venom ecology, functions and compositional variation across bioclimatic zones, partly because these nocturnal snakes are highly elusive, making it difficult to find them in the wild. We aim to address this knowledge gap by characterising the venom composition and toxicity profiles of the pan-Indian populations (n = 8) of B. caeruleus using a combination of proteomics, receptor-toxin interaction assays, biochemical experiments, pharmacological tests and preclinical evaluations. We reveal considerable variation in venom composition, functions, and pharmacological activities among the geographically distinct populations of B. caeruleus. Furthermore, toxin-receptor interaction assays provide insights into their feeding ecology and prey-predator interactions. Finally, in vitro and in vivo experiments revealed the poor neutralising potencies of Indian antivenoms towards most populations of the common krait. Our findings highlight the alarming need to develop efficacious snakebite therapy in India to treat bites from this medically most important elapid snake.
Collapse
Affiliation(s)
| | | | | | | | - Kartik Sunagar
- Evolutionary Venomics Lab, Centre for Ecological Sciences, Indian Institute of Science, Bangalore, Karnataka, India
| |
Collapse
|
172
|
Gualdrón-López M, Ayllon-Hermida A, Cortes-Serra N, Resa-Infante P, Bech-Serra JJ, Aparici-Herraiz I, Nicolau-Fernandez M, Erkizia I, Gutierrez-Chamorro L, Marfil S, Pradenas E, Ávila Nieto C, Cucurull B, Montaner-Tarbés S, Muelas M, Sotil R, Ballana E, Urrea V, Fraile L, Montoya M, Vergara J, Segales J, Carrillo J, Izquierdo-Useros N, Blanco J, Fernandez-Becerra C, de La Torre C, Pinazo MJ, Martinez-Picado J, del Portillo HA. Proteomics of circulating extracellular vesicles reveals diverse clinical presentations of COVID-19 but fails to identify viral peptides. Front Cell Infect Microbiol 2024; 14:1442743. [PMID: 39569406 PMCID: PMC11576438 DOI: 10.3389/fcimb.2024.1442743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 09/23/2024] [Indexed: 11/22/2024] Open
Abstract
Extracellular vesicles (EVs) released by virus-infected cells have the potential to encapsulate viral peptides, a characteristic that could facilitate vaccine development. Furthermore, plasma-derived EVs may elucidate pathological changes occurring in distal tissues during viral infections. We hypothesized that molecular characterization of EVs isolated from COVID-19 patients would reveal peptides suitable for vaccine development. Blood samples were collected from three cohorts: severe COVID-19 patients (G1), mild/asymptomatic cases (G2), and SARS-CoV-2-negative healthcare workers (G3). Samples were obtained at two time points: during the initial phase of the pandemic in early 2020 (m0) and eight months later (m8). Clinical data analysis revealed elevated inflammatory markers in G1. Notably, non-vaccinated individuals in G1 exhibited increased levels of neutralizing antibodies at m8, suggesting prolonged exposure to viral antigens. Proteomic profiling of EVs was performed using three distinct methods: immunocapture (targeting CD9), ganglioside-capture (utilizing Siglec-1) and size-exclusion chromatography (SEC). Contrary to our hypothesis, this analysis failed to identify viral peptides. These findings were subsequently validated through Western blot analysis targeting the RBD of the SARS-CoV-2 Spike protein's and comparative studies using samples from experimentally infected Syrian hamsters. Furthermore, analysis of the EV cargo revealed a diverse molecular profile, including components involved in the regulation of viral replication, systemic inflammation, antigen presentation, and stress responses. These findings underscore the potential significance of EVs in the pathogenesis and progression of COVID-19.
Collapse
Affiliation(s)
- Melisa Gualdrón-López
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | - Alberto Ayllon-Hermida
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | - Núria Cortes-Serra
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | - Patricia Resa-Infante
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Spain
| | | | - Iris Aparici-Herraiz
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | - Marc Nicolau-Fernandez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
| | | | | | | | | | | | | | | | | | - Ruth Sotil
- International Health Service, Hospital Clínic, Barcelona, Spain
| | - Ester Ballana
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Lorenzo Fraile
- Agro tecnio Center, Department of Animal Science, University of Lleida, Lleida, Spain
| | - Maria Montoya
- Centro de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid, Spain
| | - Julia Vergara
- Unitat Mixta d’Investigació Institute of Agrifood Research and Technology UAB (IRTA-UAB) en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Spain
- Institute of Agrifood Research and Technology (IRTA) Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Spain
| | - Joaquim Segales
- Unitat Mixta d’Investigació Institute of Agrifood Research and Technology UAB (IRTA-UAB) en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Bellaterra, Spain
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, Bellaterra, Spain
| | - Jorge Carrillo
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Julià Blanco
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Fernandez-Becerra
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Maria-Jesus Pinazo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Martinez-Picado
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- IrsiCaixa, Badalona, Spain
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- University of Vic–Central University of Catalonia (UVic-UCC), Vic, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Hernando A. del Portillo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Germans Trias I Pujol Research Institute (IGTP), Badalona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
173
|
Ballmer D, Lou HJ, Ishii M, Turk BE, Akiyoshi B. Aurora B controls anaphase onset and error-free chromosome segregation in trypanosomes. J Cell Biol 2024; 223:e202401169. [PMID: 39196069 PMCID: PMC11354203 DOI: 10.1083/jcb.202401169] [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: 01/31/2024] [Revised: 06/12/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
Kinetochores form the interface between chromosomes and spindle microtubules and are thus under tight control by a complex regulatory circuitry. The Aurora B kinase plays a central role within this circuitry by destabilizing improper kinetochore-microtubule attachments and relaying the attachment status to the spindle assembly checkpoint. Intriguingly, Aurora B is conserved even in kinetoplastids, a group of early-branching eukaryotes which possess a unique set of kinetochore proteins. It remains unclear how their kinetochores are regulated to ensure faithful chromosome segregation. Here, we show in Trypanosoma brucei that Aurora B activity controls the metaphase-to-anaphase transition through phosphorylation of the divergent Bub1-like protein KKT14. Depletion of KKT14 overrides the metaphase arrest resulting from Aurora B inhibition, while expression of non-phosphorylatable KKT14 delays anaphase onset. Finally, we demonstrate that re-targeting Aurora B to the outer kinetochore suffices to promote mitotic exit but causes extensive chromosome missegregation in anaphase. Our results indicate that Aurora B and KKT14 are involved in an unconventional circuitry controlling cell cycle progression in trypanosomes.
Collapse
Affiliation(s)
- Daniel Ballmer
- Department of Biochemistry, University of Oxford, Oxford, UK
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Hua Jane Lou
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Midori Ishii
- Department of Biochemistry, University of Oxford, Oxford, UK
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Benjamin E. Turk
- Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Bungo Akiyoshi
- Department of Biochemistry, University of Oxford, Oxford, UK
- The Wellcome Centre for Cell Biology, Institute of Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
174
|
Wang Q, Gao G, Fang F, Wang Q, Lundquist PK, Sun L. A simple and efficient approach for preparing cationic coating with tunable electroosmotic flow for capillary zone electrophoresis-mass spectrometry-based top-down proteomics. Anal Chim Acta 2024; 1328:343162. [PMID: 39266194 PMCID: PMC11404064 DOI: 10.1016/j.aca.2024.343162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/14/2024]
Abstract
BACKGROUND Capillary zone electrophoresis-tandem mass spectrometry (CZE-MS/MS) has become a valuable analytical technique in top-down proteomics (TDP). CZE-MS/MS-based TDP typically employs separation capillaries with neutral coatings (i.e., linear polyacrylamide, LPA). However, issues related to separation resolution and reproducibility remain with the LPA-coated capillaries due to the unavoidable non-specific protein adsorption onto the capillary wall. Cationic coatings can be critical alternatives to LPA coating for CZE-MS/MS-based TDP due to the electrostatic repulsion between the positively charged capillary inner wall and proteoform molecules in the acidic separation buffer. Unfortunately, there are only very few studies using cationic coating-based CZE-MS/MS for TDP studies. RESULTS In this work, we aimed to develop a simple and efficient approach for preparing separation capillaries with a cationic coating, i.e., poly (acrylamide-co-(3-acrylamidopropyl) trimethylammonium chloride [PAMAPTAC]) for CZE-MS/MS-based TDP. The PAMAPTAC coating-based CZE-MS produced significantly better separation resolution of proteoforms compared to the traditionally used LPA-coated approach. It achieved reproducible separation and measurement of a simple proteoform mixture and a complex proteome sample (i.e., a yeast cell lysate) regarding migration time, proteoform intensity, and the number of proteoform identifications. The PAMAPTAC coating-based CZE-MS enabled the detection of large proteoforms (≥30 kDa) from the yeast cell lysate reproducibly without any size-based prefractionation. Interestingly, the mobility of proteoforms using the PAMAPTAC coating can be predicted accurately using a simple semi-empirical model. SIGNIFICANCE The results render the PAMAPTAC coating as a valuable alternative to the LPA coating to advance CZE-MS-based TDP towards high-resolution separation and highly reproducible measurement of proteoforms in complex samples.
Collapse
Affiliation(s)
- Qianjie Wang
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA; Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA; Plant Resilience Institute, Michigan State University, East Lansing, MI, 48824, USA
| | - Guangyao Gao
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
| | - Fei Fang
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
| | - Qianyi Wang
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA
| | - Peter K Lundquist
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, 48824, USA; Plant Resilience Institute, Michigan State University, East Lansing, MI, 48824, USA
| | - Liangliang Sun
- Department of Chemistry, Michigan State University, 578 S Shaw Lane, East Lansing, MI, 48824, USA.
| |
Collapse
|
175
|
Meyer H, Bossen J, Janz M, Müller X, Künzel S, Roeder T, Paululat A. Combined transcriptome and proteome profiling reveal cell-type-specific functions of Drosophila garland and pericardial nephrocytes. Commun Biol 2024; 7:1424. [PMID: 39487357 PMCID: PMC11530456 DOI: 10.1038/s42003-024-07062-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024] Open
Abstract
Drosophila nephrocytes are specialised cells that share critical functional, morphological, and molecular features with mammalian podocytes. Accordingly, nephrocytes represent a preferred invertebrate model for human glomerular disease. Here, we established a method for cell-specific isolation of the two types of Drosophila nephrocytes, garland and pericardial cells, from animals of different developmental stages and ages. Mass spectrometry-based proteomics and RNA-Seq-based transcriptomics were applied to characterise the proteome and transcriptome of the respective cells in an integrated and complementary manner. We observed characteristic changes in the proteome and transcriptome due to cellular ageing. Furthermore, functional enrichment analyses suggested that larval and adult nephrocytes, as well as garland and pericardial nephrocytes, fulfil distinct physiological functions. In addition, the pericardial nephrocytes were characterised by transcriptomic and proteomic profiles suggesting an atypical energy metabolism with very low oxidative phosphorylation rates. Moreover, the nephrocytes displayed typical signatures of extensive immune signalling and showed an active antimicrobial response to an infection. Factor-specific comparisons identified novel candidate proteins either expressed and secreted by the nephrocytes or sequestered by them. The data generated in this study represent a valuable basis for a more specific application of the Drosophila model in analysing renal cell function in health and disease.
Collapse
Affiliation(s)
- Heiko Meyer
- Department of Zoology & Developmental Biology, University of Osnabrück, 49076, Osnabrück, Germany
- Center of Cellular Nanoanalytics Osnabrück-CellNanOs, 49076, Osnabrück, Germany
| | - Judith Bossen
- University of Kiel, Zoology, Molecular Physiology, 24098, Kiel, Germany
- German Center for Lung Research (DZL), Airway Research Center North (ARCN), Kiel, Germany
| | - Maren Janz
- Department of Zoology & Developmental Biology, University of Osnabrück, 49076, Osnabrück, Germany
| | - Xenia Müller
- University of Kiel, Zoology, Molecular Physiology, 24098, Kiel, Germany
| | - Sven Künzel
- Max-Planck Institute for Evolutionary Biology, Plön, Germany
| | - Thomas Roeder
- University of Kiel, Zoology, Molecular Physiology, 24098, Kiel, Germany.
- German Center for Lung Research (DZL), Airway Research Center North (ARCN), Kiel, Germany.
| | - Achim Paululat
- Department of Zoology & Developmental Biology, University of Osnabrück, 49076, Osnabrück, Germany.
- Center of Cellular Nanoanalytics Osnabrück-CellNanOs, 49076, Osnabrück, Germany.
| |
Collapse
|
176
|
Tasegian A, Dikovskaya D, Scott MM, Chawla AS, Pemberton R, Helps T, Meus T, McLean MH, Swamy M. LRRK2 is not required for lysozyme expression in Paneth cells. Nat Immunol 2024; 25:2037-2039. [PMID: 39379660 DOI: 10.1038/s41590-024-01972-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 09/06/2024] [Indexed: 10/10/2024]
Affiliation(s)
- Anna Tasegian
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
- Amphista Therapeutics, Cambridge, UK
| | - Dina Dikovskaya
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Molly M Scott
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Amanpreet Singh Chawla
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Rebecca Pemberton
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Thomas Helps
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Tosca Meus
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Mairi H McLean
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Ninewells Hospital, Dundee, UK
| | - Mahima Swamy
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
177
|
Porras MÁG, Assié A, Tietjen M, Violette M, Kleiner M, Gruber-Vodicka H, Dubilier N, Leisch N. An intranuclear bacterial parasite of deep-sea mussels expresses apoptosis inhibitors acquired from its host. Nat Microbiol 2024; 9:2877-2891. [PMID: 39242818 PMCID: PMC11521996 DOI: 10.1038/s41564-024-01808-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/13/2024] [Indexed: 09/09/2024]
Abstract
A limited number of bacteria are able to colonize the nuclei of eukaryotes. 'Candidatus Endonucleobacter' infects the nuclei of deep-sea mussels, where it replicates to ≥80,000 bacteria per nucleus and causes nuclei to swell to 50 times their original size. How these parasites are able to replicate and avoid apoptosis is not known. Dual RNA-sequencing transcriptomes of infected nuclei isolated using laser-capture microdissection revealed that 'Candidatus Endonucleobacter' does not obtain most of its nutrition from nuclear DNA or RNA. Instead, 'Candidatus Endonucleobacter' upregulates genes for importing and digesting sugars, lipids, amino acids and possibly mucin from its host. It likely prevents apoptosis of host cells by upregulating 7-13 inhibitors of apoptosis, proteins not previously seen in bacteria. Comparative phylogenetic analyses revealed that 'Ca. Endonucleobacter' acquired inhibitors of apoptosis through horizontal gene transfer from their hosts. Horizontal gene transfer from eukaryotes to bacteria is assumed to be rare, but may be more common than currently recognized.
Collapse
Affiliation(s)
| | - Adrien Assié
- Max Planck Institute for Marine Microbiology, Bremen, Germany
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Målin Tietjen
- Max Planck Institute for Marine Microbiology, Bremen, Germany
| | - Marlene Violette
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Manuel Kleiner
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Harald Gruber-Vodicka
- Max Planck Institute for Marine Microbiology, Bremen, Germany
- Zoological Institute, Christian-Albrechts-University, Kiel, Germany
| | - Nicole Dubilier
- Max Planck Institute for Marine Microbiology, Bremen, Germany.
| | - Nikolaus Leisch
- Max Planck Institute for Marine Microbiology, Bremen, Germany.
- European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
178
|
Novotna S, Maia LA, Radaszkiewicz KA, Roudnicky P, Harnos J. Linking planar polarity signalling to actomyosin contractility during vertebrate neurulation. Open Biol 2024; 14:240251. [PMID: 39561813 PMCID: PMC11576107 DOI: 10.1098/rsob.240251] [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: 09/05/2024] [Revised: 09/19/2024] [Accepted: 10/02/2024] [Indexed: 11/21/2024] Open
Abstract
Actomyosin contractility represents an ancient feature of eukaryotic cells participating in many developmental and homeostasis events, including tissue morphogenesis, muscle contraction and cell migration, with dysregulation implicated in various pathological conditions, such as cancer. At the molecular level, actomyosin comprises actin bundles and myosin motor proteins that are sensitive to posttranslational modifications like phosphorylation. While the molecular components of actomyosin are well understood, the coordination of contractility by extracellular and intracellular signals, particularly from cellular signalling pathways, remains incompletely elucidated. This study focuses on WNT/planar cell polarity (PCP) signalling, previously associated with actomyosin contractility during vertebrate neurulation. Our investigation reveals that the main cytoplasmic PCP proteins, Prickle and Dishevelled, interact with key actomyosin components such as myosin light chain 9 (MLC9), leading to its phosphorylation and localized activation. Using proteomics and microscopy approaches, we demonstrate that both PCP proteins actively control actomyosin contractility through Rap1 small GTPases in relevant in vitro and in vivo models. These findings unveil a novel mechanism of how PCP signalling regulates actomyosin contractility through MLC9 and Rap1 that is relevant to vertebrate neurulation.
Collapse
Affiliation(s)
- Sarka Novotna
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno62500, Czechia
| | - Lorena Agostini Maia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno62500, Czechia
| | | | - Pavel Roudnicky
- CEITEC-Central European Institute of Technology, Masaryk University, Brno62500, Czechia
- National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno62500, Czechia
| | - Jakub Harnos
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno62500, Czechia
| |
Collapse
|
179
|
Adriaenssens E, Nguyen TN, Sawa-Makarska J, Khuu G, Schuschnig M, Shoebridge S, Skulsuppaisarn M, Watts EM, Csalyi KD, Padman BS, Lazarou M, Martens S. Control of mitophagy initiation and progression by the TBK1 adaptors NAP1 and SINTBAD. Nat Struct Mol Biol 2024; 31:1717-1731. [PMID: 38918639 PMCID: PMC11564117 DOI: 10.1038/s41594-024-01338-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 05/22/2024] [Indexed: 06/27/2024]
Abstract
Mitophagy preserves overall mitochondrial fitness by selectively targeting damaged mitochondria for degradation. The regulatory mechanisms that prevent PTEN-induced putative kinase 1 (PINK1) and E3 ubiquitin ligase Parkin (PINK1/Parkin)-dependent mitophagy and other selective autophagy pathways from overreacting while ensuring swift progression once initiated are largely elusive. Here, we demonstrate how the TBK1 (TANK-binding kinase 1) adaptors NAP1 (NAK-associated protein 1) and SINTBAD (similar to NAP1 TBK1 adaptor) restrict the initiation of OPTN (optineurin)-driven mitophagy by competing with OPTN for TBK1. Conversely, they promote the progression of nuclear dot protein 52 (NDP52)-driven mitophagy by recruiting TBK1 to NDP52 and stabilizing its interaction with FIP200. Notably, OPTN emerges as the primary recruiter of TBK1 during mitophagy initiation, which in return boosts NDP52-mediated mitophagy. Our results thus define NAP1 and SINTBAD as cargo receptor rheostats, elevating the threshold for mitophagy initiation by OPTN while promoting the progression of the pathway once set in motion by supporting NDP52. These findings shed light on the cellular strategy to prevent pathway hyperactivity while still ensuring efficient progression.
Collapse
Affiliation(s)
- Elias Adriaenssens
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria.
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Vienna, Austria.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Thanh Ngoc Nguyen
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Justyna Sawa-Makarska
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Vienna, Austria
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Grace Khuu
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Martina Schuschnig
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Vienna, Austria
| | - Stephen Shoebridge
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Vienna, Austria
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Marvin Skulsuppaisarn
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - Emily Maria Watts
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
| | - Kitti Dora Csalyi
- Max Perutz Labs BioOptics FACS Facility, Max Perutz Labs, University of Vienna, Vienna BioCenter Campus (VBC), Vienna, Austria
| | - Benjamin Scott Padman
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia
- Harry Perkins Institute of Medical Research, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, Western Australia, Australia
| | - Michael Lazarou
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia.
| | - Sascha Martens
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria.
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Vienna, Austria.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
180
|
Zuniga NR, Frost DC, Kuhn K, Shin M, Whitehouse RL, Wei TY, He Y, Dawson SL, Pike I, Bomgarden RD, Gygi SP, Paulo JA. Achieving a 35-Plex Tandem Mass Tag Reagent Set through Deuterium Incorporation. J Proteome Res 2024; 23:5153-5165. [PMID: 39380184 DOI: 10.1021/acs.jproteome.4c00668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Mass spectrometry-based sample multiplexing with isobaric tags permits the development of high-throughput and precise quantitative biological assays with proteome-wide coverage and minimal missing values. Here, we nearly doubled the multiplexing capability of the TMTpro reagent set to a 35-plex through the incorporation of one deuterium isotope into the reporter group. Substituting deuterium frequently results in suboptimal peak coelution, which can compromise the accuracy of reporter ion-based quantification. To counteract the deuterium effect on quantitation, we implemented a strategy that necessitated the segregation of nondeuterium and deuterium-containing channels into distinct subplexes during normalization procedures, with reassembly through a common bridge channel. This multiplexing strategy of "design independent sub-plexes but acquire together" (DISAT) was used to compare protein expression differences between human cell lines and in a cysteine-profiling (i.e., chemoproteomics) experiment to identify compounds binding to cysteine-113 of Pin1.
Collapse
Affiliation(s)
- Nathan R Zuniga
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Dustin C Frost
- Thermo Fisher Scientific, Rockford, Illinois 61101, United States
| | | | - Myungsun Shin
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Rebecca L Whitehouse
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ting-Yu Wei
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Yuchen He
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Shane L Dawson
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ian Pike
- Proteome Sciences, London KT15 2HJ, U.K
| | - Ryan D Bomgarden
- Thermo Fisher Scientific, Rockford, Illinois 61101, United States
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
181
|
Ramarajan MG, Parthasarathy KTS, Gaikwad KB, Joshi N, Garapati K, Kandasamy RK, Sharma J, Pandey A. Alterations in Hurler-Scheie Syndrome Revealed by Mass Spectrometry-Based Proteomics and Phosphoproteomics Analysis. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2024; 28:548-562. [PMID: 39469785 DOI: 10.1089/omi.2024.0171] [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: 10/30/2024]
Abstract
Hurler-Scheie syndrome (MPS IH/S), also known as mucopolysaccharidosis type I-H/S (MPS IH/S), is a lysosomal storage disorder caused by deficiency of the enzyme alpha-L-iduronidase (IDUA) leading to the accumulation of glycosaminoglycans (GAGs) in various tissues, resulting in a wide range of symptoms affecting different organ systems. Postgenomic omics technologies offer the promise to understand the changes in proteome, phosphoproteome, and phosphorylation-based signaling in MPS IH/S. Accordingly, we report here a large dataset and the proteomic and phosphoproteomic analyses of fibroblasts derived from patients with MPS IH/S (n = 8) and healthy individuals (n = 8). We found that protein levels of key lysosomal enzymes such as cathepsin D, prosaposin, arylsulfatases (arylsulfatase A and arylsulfatase B), and IDUA were downregulated. We identified 16,693 unique phosphopeptides, corresponding to 4,605 proteins, in patients with MPS IH/S. We found that proteins related to the cell cycle, mitotic spindle assembly, apoptosis, and cytoskeletal organization were differentially phosphorylated in MPS IH/S. We identified 12 kinases that were differentially phosphorylated, including hyperphosphorylation of cyclin-dependent kinases 1 and 2, hypophosphorylation of myosin light chain kinase, and calcium/calmodulin-dependent protein kinases. Taken together, the findings of the present study indicate significant alterations in proteins involved in cytoskeletal changes, cellular dysfunction, and apoptosis. These new observations significantly contribute to the current understanding of the pathophysiology of MPS IH/S specifically, and the molecular mechanisms involved in the storage of GAGs in MPS more generally. Further translational clinical omics studies are called for to pave the way for diagnostics and therapeutics innovation for patients with MPS IH/S.
Collapse
Affiliation(s)
- Madan Gopal Ramarajan
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - K T Shreya Parthasarathy
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
| | - Kiran Bharat Gaikwad
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
| | - Neha Joshi
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kishore Garapati
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Richard K Kandasamy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota, USA
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jyoti Sharma
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
182
|
Sweatt AJ, Griffiths CD, Groves SM, Paudel BB, Wang L, Kashatus DF, Janes KA. Proteome-wide copy-number estimation from transcriptomics. Mol Syst Biol 2024; 20:1230-1256. [PMID: 39333715 PMCID: PMC11535397 DOI: 10.1038/s44320-024-00064-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/29/2024] Open
Abstract
Protein copy numbers constrain systems-level properties of regulatory networks, but proportional proteomic data remain scarce compared to RNA-seq. We related mRNA to protein statistically using best-available data from quantitative proteomics and transcriptomics for 4366 genes in 369 cell lines. The approach starts with a protein's median copy number and hierarchically appends mRNA-protein and mRNA-mRNA dependencies to define an optimal gene-specific model linking mRNAs to protein. For dozens of cell lines and primary samples, these protein inferences from mRNA outmatch stringent null models, a count-based protein-abundance repository, empirical mRNA-to-protein ratios, and a proteogenomic DREAM challenge winner. The optimal mRNA-to-protein relationships capture biological processes along with hundreds of known protein-protein complexes, suggesting mechanistic relationships. We use the method to identify a viral-receptor abundance threshold for coxsackievirus B3 susceptibility from 1489 systems-biology infection models parameterized by protein inference. When applied to 796 RNA-seq profiles of breast cancer, inferred copy-number estimates collectively re-classify 26-29% of luminal tumors. By adopting a gene-centered perspective of mRNA-protein covariation across different biological contexts, we achieve accuracies comparable to the technical reproducibility of contemporary proteomics.
Collapse
Affiliation(s)
- Andrew J Sweatt
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Cameron D Griffiths
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Sarah M Groves
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - B Bishal Paudel
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lixin Wang
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | - David F Kashatus
- Department of Microbiology, Immunology & Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Kevin A Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA.
- Department of Biochemistry & Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
183
|
Raabe J, Wittig I, Laurette P, Stathopoulou K, Brand T, Schulze T, Klampe B, Orthey E, Cabrera-Orefice A, Meisterknecht J, Thiemann E, Laufer SD, Shibamiya A, Reinsch M, Fuchs S, Kaiser J, Yang J, Zehr S, Wrona KM, Lorenz K, Lukowski R, Hansen A, Gilsbach R, Brandes RP, Ulmer BM, Eschenhagen T, Cuello F. Physioxia rewires mitochondrial complex composition to protect stem cell viability. Redox Biol 2024; 77:103352. [PMID: 39341035 PMCID: PMC11466565 DOI: 10.1016/j.redox.2024.103352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) are an invaluable tool to study molecular mechanisms on a human background. Culturing stem cells at an oxygen level different from their microenvironmental niche impacts their viability. To understand this mechanistically, dermal skin fibroblasts of 52 probands were reprogrammed into hiPSCs, followed by either hyperoxic (20 % O2) or physioxic (5 % O2) culture and proteomic profiling. Analysis of chromosomal stability by Giemsa-banding revealed that physioxic -cultured hiPSC clones exhibited less pathological karyotypes than hyperoxic (e.g. 6 % vs. 32 % mosaicism), higher pluripotency as evidenced by higher Stage-Specific Embryonic Antigen 3 positivity, higher glucose consumption and lactate production. Global proteomic analysis demonstrated lower abundance of several subunits of NADH:ubiquinone oxidoreductase (complex I) and an underrepresentation of pathways linked to oxidative phosphorylation and cellular senescence. Accordingly, release of the pro-senescent factor IGFBP3 and β-galactosidase staining were lower in physioxic hiPSCs. RNA- and ATAC-seq profiling revealed a distinct hypoxic transcription factor-binding footprint, amongst others higher expression of the HIF1α-regulated target NDUFA4L2 along with increased chromatin accessibility of the NDUFA4L2 gene locus. While mitochondrial DNA content did not differ between groups, physioxic hiPSCs revealed lower polarized mitochondrial membrane potential, altered mitochondrial network appearance and reduced basal respiration and electron transfer capacity. Blue-native polyacrylamide gel electrophoresis coupled to mass spectrometry of the mitochondrial complexes detected higher abundance of NDUFA4L2 and ATP5IF1 and loss of incorporation into complex IV or V, respectively. Taken together, physioxic culture of hiPSCs improved chromosomal stability, which was associated with downregulation of oxidative phosphorylation and senescence and extensive re-wiring of mitochondrial complex composition.
Collapse
Affiliation(s)
- Janice Raabe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ilka Wittig
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany
| | - Patrick Laurette
- Institute of Experimental Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Konstantina Stathopoulou
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Theresa Brand
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
| | - Thomas Schulze
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Birgit Klampe
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ellen Orthey
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Alfredo Cabrera-Orefice
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Jana Meisterknecht
- Functional Proteomics Center, Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Ellen Thiemann
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Sandra D Laufer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Aya Shibamiya
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Marina Reinsch
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Sigrid Fuchs
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jennifer Kaiser
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Jiaqi Yang
- Institute of Pharmacy, Experimental Pharmacology, University Tübingen, 72076 Tübingen, Germany
| | - Simonida Zehr
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany; Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Kinga M Wrona
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Kristina Lorenz
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany; Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | - Robert Lukowski
- Institute of Pharmacy, Experimental Pharmacology, University Tübingen, 72076 Tübingen, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Ralf Gilsbach
- Institute of Experimental Cardiology, Heidelberg University Hospital, 69120 Heidelberg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany
| | - Ralf P Brandes
- DZHK (German Center for Cardiovascular Research), Partner Site Rhein-Main, Frankfurt, Germany; Institute for Cardiovascular Physiology, Goethe-University, 60590 Frankfurt am Main, Germany
| | - Bärbel M Ulmer
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| | - Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany.
| |
Collapse
|
184
|
Moreno-Ulloa A, Zárate-Córdova VL, Ramírez-Sánchez I, Cruz-López JC, Perez-Ortiz A, Villarreal-Garza C, Díaz-Chávez J, Estrada-Mena B, Antonio-Aguirre B, López-Almanza PX, Lira-Romero E, Estrada-Mena FJ. Evaluation of a Proteomics-Guided Protein Signature for Breast Cancer Detection in Breast Tissue. J Proteome Res 2024; 23:4907-4923. [PMID: 39412830 DOI: 10.1021/acs.jproteome.4c00295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
The distinction between noncancerous and cancerous breast tissues is challenging in clinical settings, and discovering new proteomics-based biomarkers remains underexplored. Through a pilot proteomic study (discovery cohort), we aimed to identify a protein signature indicative of breast cancer for subsequent validation using six published proteomics/transcriptomics data sets (validation cohorts). Sequential window acquisition of all theoretical (SWATH)-based mass spectrometry revealed 370 differentially abundant proteins between noncancerous tissue and breast cancer. Protein-protein interaction-based networks and enrichment analyses revealed dysregulation in pathways associated with extracellular matrix organization, platelet degranulation, the innate immune system, and RNA metabolism in breast cancer. Through multivariate unsupervised analysis, we identified a four-protein signature (OGN, LUM, DCN, and COL14A1) capable of distinguishing breast cancer. This dysregulation pattern was consistently verified across diverse proteomics and transcriptomics data sets. Dysregulation magnitude was notably higher in poor-prognosis breast cancer subtypes like Basal-Like and HER2 compared to Luminal A. Diagnostic evaluation (receiver operating characteristic (ROC) curves) of the signature in distinguishing breast cancer from noncancerous tissue revealed area under the curve (AUC) ranging from 0.87 to 0.9 with predictive accuracy of 80% to 82%. Upon stratifying, to solely include the Basal-Like/Triple-Negative subtype, the ROC AUC increased to 0.922-0.959 with predictive accuracy of 84.2%-89%. These findings suggest a potential role for the identified signature in distinguishing cancerous from noncancerous breast tissue, offering insights into enhancing diagnostic accuracy.
Collapse
Affiliation(s)
- Aldo Moreno-Ulloa
- Laboratorio MS2, Departamento de Innovación Biomédica, CICESE, Ensenada 22860, Baja California, México
| | - Vareska L Zárate-Córdova
- Laboratorio MS2, Departamento de Innovación Biomédica, CICESE, Ensenada 22860, Baja California, México
- Posgrado en Ciencias de la Vida, CICESE, Ensenada 22860, Baja California, México
| | - Israel Ramírez-Sánchez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, IPN, Ciudad de México 11340, México
| | - Juan Carlos Cruz-López
- Hospital Puebla, Puebla 72197, Pue., México
- Hospital General Zona Norte SSEP, Puebla 72200, Pue., México
| | - Andric Perez-Ortiz
- Escuela de Medicina, Universidad Panamericana, Ciudad de México 03920, México
- Departamento de Cirugía, Centro Médico ABC, Ciudad de México 05348, México
| | - Cynthia Villarreal-Garza
- Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, Monterrey 66260, Nuevo León, México
| | - José Díaz-Chávez
- Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, UNAM/Instituto Nacional de Cancerología, Ciudad de México 14080, México
| | - Benito Estrada-Mena
- Escuela de Enfermería, Universidad Panamericana, Ciudad de México 03920, México
- Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | | | | | | | | |
Collapse
|
185
|
Salovska B, Li W, Bernhardt OM, Germain PL, Gandhi T, Reiter L, Liu Y. A Comprehensive and Robust Multiplex-DIA Workflow Profiles Protein Turnover Regulations Associated with Cisplatin Resistance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620709. [PMID: 39554001 PMCID: PMC11565775 DOI: 10.1101/2024.10.28.620709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Measuring protein turnover is essential for understanding cellular biological processes and advancing drug discovery. The multiplex DIA mass spectrometry (DIA-MS) approach, combined with dynamic SILAC labeling (pulse-SILAC, or pSILAC), has proven to be a reliable method for analyzing protein turnover and degradation kinetics. Previous multiplex DIA-MS workflows have employed various strategies, including leveraging the highest isotopic labeling channels of peptides to enhance the detection of isotopic MS signal pairs or clusters. In this study, we introduce an improved and robust workflow that integrates a novel machine learning strategy and channel-specific statistical filtering, enabling dynamic adaptation to systematic or temporal variations in channel ratios. This allows comprehensive profiling of protein turnover throughout the pSILAC experiment without relying solely on the highest channel signals. Additionally, we developed KdeggeR , a data processing and analysis package optimized for pSILAC-DIA experiments, which estimates and visualizes peptide and protein degradation rates and dynamic profiles. Our integrative workflow was benchmarked on both 2-channel and 3-channel standard DIA datasets and across two mass spectrometry platforms, demonstrating its broad applicability. Finally, applying this workflow to an aneuploid cancer cell model before and after cisplatin resistance development demonstrated a strong negative correlation between transcript regulation and protein degradation for major protein complex subunits. We also identified specific protein turnover signatures associated with cisplatin resistance.
Collapse
Affiliation(s)
- Barbora Salovska
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | - Wenxue Li
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
| | | | - Pierre-Luc Germain
- Institute for Neuroscience, Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | | | | | - Yansheng Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
- Cancer Biology Institute, Yale University School of Medicine, West Haven, CT 06516, USA
- Department of Biomedical Informatics & Data Science, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
186
|
Rossio V, Paulo JA. Comparative Proteome-Wide Abundance Profiling of Yeast Strains Deleted for Cdc48 Adaptors. Proteomes 2024; 12:31. [PMID: 39585118 PMCID: PMC11587464 DOI: 10.3390/proteomes12040031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
The yeast ATPase Cdc48 (known as p97/VCP in human cells) plays an important role in the Ubiquitin Proteasome System. VCP is essential for cancer cell proliferation, and its dysregulation has been implicated in several neurodegenerative diseases. Cdc48 functions by extracting ubiquitylated proteins from membranes, protein complexes and chromatin by often facilitating their proteasomal degradation. Specific adaptors or cofactors, primarily belonging to the UBX domain-containing protein family (which has seven members in Saccharomyces cerevisiae) recruit Cdc48 to ubiquitylated proteins. Here, we employed sample multiplexing-based quantitative mass spectrometry to profile global protein abundance in p97 adaptor deletion strains, specifically comparing seven single deletion strains of UBX domain-containing proteins and the Cuz1 deletion strain, which belongs to the zinc finger AN1-type domain protein family. We observed that each strain showed unique sets of differentially abundant proteins compared to the wild type. Our analysis also revealed a role for Ubx3 in maintaining wild type levels of mitochondrial proteins. Overall, we identified ~1400 differentially abundant proteins in the absence of a specific Cdc48 adaptor. This unique dataset offers a valuable resource for studying the functions of these adaptors, aiming to achieve a better understanding of the cellular processes regulated by Cdc48 itself and to deepen our understanding of the Ubiquitin Proteasome System.
Collapse
Affiliation(s)
- Valentina Rossio
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
187
|
Santa C, Rodrigues JE, Martinho A, Mendes VM, Madeira N, Coroa M, Santos V, Morais S, Bajouco M, Costa H, Anjo SI, Baldeiras I, Macedo A, Manadas B. Proteomic analysis of peripheral blood mononuclear cells in first episode psychosis - Protein and peptide-centered approaches to elucidate potential diagnostic biomarkers. J Proteomics 2024; 309:105296. [PMID: 39218299 DOI: 10.1016/j.jprot.2024.105296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Diagnosing patients suffering from psychotic disorders is far from being achieved with molecular support, despite all the efforts to study these disorders from different perspectives. Characterizing the proteome of easily obtainable blood specimens, such as the peripheral blood mononuclear cells (PBMCs), has particular interest in biomarker discovery and generating pathophysiological knowledge. This approach has been explored in psychiatry, and while generating valuable information, it has not translated into meaningful biomarker discovery. In this project, we report the proof-of-concept of a methodology that aims to explore further information obtained with classical proteomics approaches that is easily overlooked. PBMC samples from first-episode psychosis and control subjects were subjected to a SWATH-MS approach, and the classical protein relative quantification was performed, where 389 proteins were found to be important to distinguish the two groups. Individual analysis of the quantified peptides was also performed, highlighting peptides of unchanged proteins that were significantly altered. With the combination of protein- and peptide-centered proteomics approaches, it is possible to highlight that information about proteoforms, namely regulation at the peptide level possibly due to post-translational modifications, is routinely overlooked and that its diagnostic potential should be further explored. SIGNIFICANCE: Our exploratory findings highlight the potential of MS-based proteomics strategies, combining protein- and peptide-centered approaches, to aid clinical decision-making in first-episode psychosis, helping to establish early biomarkers for schizophrenia and other psychotic disorders. Particularly, the less popular peptide-centered approach allows the identification/measurement of overlooked modulated peptides that may have potential biomarker characteristics. The application in parallel of protein- and peptide-centered strategies is transversal to research of other diseases, potentially allowing a more comprehensive characterization of the metabolic/pathophysiological alterations related to a specific disease.
Collapse
Affiliation(s)
- Catia Santa
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - João E Rodrigues
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Ana Martinho
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Vera M Mendes
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Nuno Madeira
- Faculty of Medicine of the University of Coimbra, University of Coimbra, Portugal; Psychiatry Department, Centro Hospitalar e Universitário de Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Portugal
| | - Manuel Coroa
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine of the University of Coimbra, University of Coimbra, Portugal; Psychiatry Department, Centro Hospitalar e Universitário de Coimbra, Portugal
| | - Vítor Santos
- CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine of the University of Coimbra, University of Coimbra, Portugal; Psychiatry Department, Centro Hospitalar e Universitário de Coimbra, Portugal
| | - Sofia Morais
- Faculty of Medicine of the University of Coimbra, University of Coimbra, Portugal; Psychiatry Department, Centro Hospitalar e Universitário de Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Portugal
| | - Miguel Bajouco
- Faculty of Medicine of the University of Coimbra, University of Coimbra, Portugal; Psychiatry Department, Centro Hospitalar e Universitário de Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Portugal
| | - Hélder Costa
- Psychiatry Department, Centro Hospitalar e Universitário de Coimbra, Portugal
| | - Sandra I Anjo
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine of the University of Coimbra, University of Coimbra, Portugal
| | - Antonio Macedo
- Faculty of Medicine of the University of Coimbra, University of Coimbra, Portugal; Psychiatry Department, Centro Hospitalar e Universitário de Coimbra, Portugal; CIBIT - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, Portugal.
| | - Bruno Manadas
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; III Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Portugal.
| |
Collapse
|
188
|
Jin H, Yeom JH, Shin E, Ha Y, Liu H, Kim D, Joo M, Kim YH, Kim HK, Ryu M, Kim HM, Kim J, Kim KP, Hahn Y, Bae J, Lee K. 5'-tRNA Gly(GCC) halves generated by IRE1α are linked to the ER stress response. Nat Commun 2024; 15:9273. [PMID: 39468069 PMCID: PMC11519470 DOI: 10.1038/s41467-024-53624-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/16/2024] [Indexed: 10/30/2024] Open
Abstract
Transfer RNA halves (tRHs) have various biological functions. However, the biogenesis of specific 5'-tRHs under certain conditions remains unknown. Here, we report that inositol-requiring enzyme 1α (IRE1α) cleaves the anticodon stem-loop region of tRNAGly(GCC) to produce 5'-tRHs (5'-tRH-GlyGCC) with highly selective target discrimination upon endoplasmic reticulum (ER) stress. Levels of 5'-tRH-GlyGCC positively affect cancer cell proliferation and modulate mRNA isoform biogenesis both in vitro and in vivo; these effects require co-expression of two nuclear ribonucleoproteins, HNRNPM and HNRNPH2, which we identify as binding proteins of 5'-tRH-GlyGCC. In addition, under ER stress in vivo, we observe simultaneous induction of IRE1α and 5'-tRH-GlyGCC expression in mouse organs and a distantly related organism, Cryptococcus neoformans. Thus, collectively, our findings indicate an evolutionarily conserved function for IRE1α-generated 5'-tRH-GlyGCC in cellular adaptation upon ER stress.
Collapse
Affiliation(s)
- Hanyong Jin
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002, Jilin, China
| | - Ji-Hyun Yeom
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
- R & D Institute, NES Biotechnology, Seoul, 06974, Republic of Korea
| | - Eunkyoung Shin
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
- Department of Microbiology, School of Medicine, Catholic University of Daegu, Daegu, 42472, Republic of Korea
| | - Yoonjie Ha
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Haifeng Liu
- School of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Daeyoung Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Minju Joo
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
- R & D Institute, NES Biotechnology, Seoul, 06974, Republic of Korea
| | - Yong-Hak Kim
- Department of Microbiology, School of Medicine, Catholic University of Daegu, Daegu, 42472, Republic of Korea
| | - Hak Kyun Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Minkyung Ryu
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
- R & D Institute, NES Biotechnology, Seoul, 06974, Republic of Korea
| | - Hong-Man Kim
- R & D Institute, NES Biotechnology, Seoul, 06974, Republic of Korea
| | - Jeongkyu Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Keun P Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yoonsoo Hahn
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jeehyeon Bae
- School of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| | - Kangseok Lee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
- R & D Institute, NES Biotechnology, Seoul, 06974, Republic of Korea.
| |
Collapse
|
189
|
Katare PB, Dalmao-Fernandez A, Mengeste AM, Navabakbar F, Hamarsland H, Ellefsen S, Berge RK, Bakke HG, Nyman TA, Kase ET, Rustan AC, Thoresen GH. Krill oil supplementation in vivo promotes increased fuel metabolism and protein synthesis in cultured human skeletal muscle cells. Front Nutr 2024; 11:1452768. [PMID: 39555189 PMCID: PMC11565515 DOI: 10.3389/fnut.2024.1452768] [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: 06/21/2024] [Accepted: 10/07/2024] [Indexed: 11/19/2024] Open
Abstract
Introduction Krill oil is a dietary supplement derived from Antarctic krill; a small crustacean found in the ocean. Krill oil is a rich source of omega-3 fatty acids, specifically eicosapentaenoic acid and docosahexaenoic acid, as well as the antioxidant astaxanthin. The aim of this study was to investigate the effects of krill oil supplementation, compared to placebo oil (high oleic sunflower oil added astaxanthin), in vivo on energy metabolism and substrate turnover in human skeletal muscle cells. Methods Skeletal muscle cells (myotubes) were obtained before and after a 7-week krill oil or placebo oil intervention, and glucose and oleic acid metabolism and leucine accumulation, as well as effects of different stimuli in vitro, were studied in the myotubes. The functional data were combined with proteomic and transcriptomic analyses. Results In vivo intervention with krill oil increased oleic acid oxidation and leucine accumulation in skeletal muscle cells, however no effects were observed on glucose metabolism. The krill oil-intervention-induced increase in oleic acid oxidation correlated negatively with changes in serum low-density lipoprotein (LDL) concentration. In addition, myotubes were also exposed to krill oil in vitro. The in vitro study revealed that 24 h of krill oil treatment increased both glucose and oleic acid metabolism in myotubes, enhancing energy substrate utilization. Transcriptomic analysis comparing myotubes obtained before and after krill oil supplementation identified differentially expressed genes associated with e.g., glycolysis/gluconeogenesis, metabolic pathways and calcium signaling pathway, while proteomic analysis demonstrated upregulation of e.g., LDL-receptor in myotubes obtained after the krill oil intervention. Conclusion These findings suggest that krill oil intervention promotes increased fuel metabolism and protein synthesis in human skeletal muscle cells, with potential implications for metabolic health.
Collapse
Affiliation(s)
- Parmeshwar B. Katare
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Andrea Dalmao-Fernandez
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Abel M. Mengeste
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Farnaz Navabakbar
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Håvard Hamarsland
- Section for Health and Exercise Physiology, Faculty of Social and Health Sciences, Inland Norway University of Applied Sciences, Lillehammer, Norway
| | - Stian Ellefsen
- Section for Health and Exercise Physiology, Faculty of Social and Health Sciences, Inland Norway University of Applied Sciences, Lillehammer, Norway
- Innlandet Hospital Trust, Lillehammer, Norway
| | - Rolf K. Berge
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Sciences, University of Bergen, Bergen, Norway
| | - Hege G. Bakke
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Tuula Anneli Nyman
- Department of Immunology, Institute of Clinical Medicine, Oslo University Hospital, University of Oslo, Oslo, Norway
| | - Eili Tranheim Kase
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - Arild C. Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
| | - G. Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Oslo, Norway
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
190
|
Jurado-Martín I, Tomás-Cortázar J, Hou Y, Sainz-Mejías M, Mysior MM, Sadonès O, Huebner J, Romero-Saavedra F, Simpson JC, Baugh JA, McClean S. Proteomic approach to identify host cell attachment proteins provides protective Pseudomonas aeruginosa vaccine antigen FtsZ. NPJ Vaccines 2024; 9:204. [PMID: 39468053 PMCID: PMC11519640 DOI: 10.1038/s41541-024-00994-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
Pseudomonas aeruginosa is an opportunistic Gram-negative pathogen that causes severe nosocomial infections in susceptible individuals due to the emergence of multidrug-resistant strains. There are no approved vaccines against P. aeruginosa infections nor candidates in active clinical development, highlighting the need for novel candidates and strategies. Using a cell-blot proteomic approach, we reproducibly identified 49 proteins involved in interactions with human lung epithelial cells across four P. aeruginosa strains. Among these were cell division protein FtsZ and outer membrane protein OpmH. Escherichia coli BL21 cells overexpressing recombinant FtsZ or rOpmH showed a 66- and 15-fold increased ability to attach to 16HBE14o- cells, further supporting their involvement in host cell attachment. Both antigens led to proliferation of NK and CD8+ cytotoxic T cells, significant increases in the production of IFN-γ, IL-17A, TNF and IL-4 in immunised mice and elicited strong antigen-specific serological IgG1 and IgG2c responses. Immunisation with FtsZ significantly reduced bacterial burden in the lungs by 1.9-log CFU and dissemination to spleen by 1.8-log CFU. The protective antigen candidate, FtsZ, would not have been identified by traditional approaches relying on either virulence mechanisms or sequence-based predictions, opening new avenues in the development of an anti-P. aeruginosa vaccine.
Collapse
Affiliation(s)
- Irene Jurado-Martín
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Julen Tomás-Cortázar
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Yueran Hou
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Maite Sainz-Mejías
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
| | - Margaritha M Mysior
- Cell Screening Laboratory, School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - Océane Sadonès
- Division of Pediatric Infectious Disease, Hauner Children's Hospital, LMU, Munich, Germany
| | - Johannes Huebner
- Division of Pediatric Infectious Disease, Hauner Children's Hospital, LMU, Munich, Germany
| | - Felipe Romero-Saavedra
- Division of Pediatric Infectious Disease, Hauner Children's Hospital, LMU, Munich, Germany
| | - Jeremy C Simpson
- Cell Screening Laboratory, School of Biology and Environmental Science, University College Dublin, Dublin, Ireland
| | - John A Baugh
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, Conway Institute, University College Dublin, Dublin, Ireland
| | - Siobhán McClean
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.
- UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin, Ireland.
| |
Collapse
|
191
|
Desai H, Andrews KH, Bergersen KV, Ofori S, Yu F, Shikwana F, Arbing MA, Boatner LM, Villanueva M, Ung N, Reed EF, Nesvizhskii AI, Backus KM. Chemoproteogenomic stratification of the missense variant cysteinome. Nat Commun 2024; 15:9284. [PMID: 39468056 PMCID: PMC11519605 DOI: 10.1038/s41467-024-53520-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/15/2024] [Indexed: 10/30/2024] Open
Abstract
Cancer genomes are rife with genetic variants; one key outcome of this variation is widespread gain-of-cysteine mutations. These acquired cysteines can be both driver mutations and sites targeted by precision therapies. However, despite their ubiquity, nearly all acquired cysteines remain unidentified via chemoproteomics; identification is a critical step to enable functional analysis, including assessment of potential druggability and susceptibility to oxidation. Here, we pair cysteine chemoproteomics-a technique that enables proteome-wide pinpointing of functional, redox sensitive, and potentially druggable residues-with genomics to reveal the hidden landscape of cysteine genetic variation. Our chemoproteogenomics platform integrates chemoproteomic, whole exome, and RNA-seq data, with a customized two-stage false discovery rate (FDR) error controlled proteomic search, which is further enhanced with a user-friendly FragPipe interface. Chemoproteogenomics analysis reveals that cysteine acquisition is a ubiquitous feature of both healthy and cancer genomes that is further elevated in the context of decreased DNA repair. Reference cysteines proximal to missense variants are also found to be pervasive, supporting heretofore untapped opportunities for variant-specific chemical probe development campaigns. As chemoproteogenomics is further distinguished by sample-matched combinatorial variant databases and is compatible with redox proteomics and small molecule screening, we expect widespread utility in guiding proteoform-specific biology and therapeutic discovery.
Collapse
Affiliation(s)
- Heta Desai
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Katrina H Andrews
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Kristina V Bergersen
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Samuel Ofori
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Flowreen Shikwana
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Mark A Arbing
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- UCLA-DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, CA, USA
| | - Lisa M Boatner
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA
| | - Miranda Villanueva
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
| | - Nicholas Ung
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Elaine F Reed
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Keriann M Backus
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, USA.
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA.
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, USA.
- UCLA-DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
192
|
Contreras W, Groenendyk J, Gentzel M, Schönberg PY, Buchholz F, Michalak M, Schröder B, Mentrup T. Selective regulation of aspartyl intramembrane protease activity by calnexin. Cell Mol Life Sci 2024; 81:441. [PMID: 39460794 PMCID: PMC11513070 DOI: 10.1007/s00018-024-05478-8] [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: 06/12/2024] [Revised: 09/09/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024]
Abstract
Signal peptide peptidase-like 2c (SPPL2c) is a testis-specific aspartyl intramembrane protease that contributes to male gamete function both by catalytic and non-proteolytic mechanisms. Here, we provide an unbiased characterisation of the in vivo interactome of SPPL2c identifying the ER chaperone calnexin as novel binding partner of this enzyme. Recruitment of calnexin specifically required the N-glycosylation within the N-terminal protease-associated domain of SPPL2c. Importantly, mutation of the single glycosylation site of SPPL2c or loss of calnexin expression completely prevented SPPL2c-mediated intramembrane proteolysis of all tested substrates. By contrast and despite rather promiscuous binding of calnexin to other SPP/SPPL proteases, expression of the chaperone was exclusively required for SPPL2c-mediated proteolysis. Despite some impact on the stability of SPPL2c most presumably due to assistance in folding of the luminal domain of the protease, calnexin appeared to be recruited rather constitutively to the protease thereby boosting its catalytic activity. In summary, we describe a novel, highly specific mode of intramembrane protease regulation, highlighting the need to systematically approach control mechanisms governing the proteolytic activity of other members of the aspartyl intramembrane protease family.
Collapse
Affiliation(s)
- Whendy Contreras
- Institute of Physiological Chemistry, Medizinische Fakultät und Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Marc Gentzel
- Core Facility Molecular Analysis - Mass Spectrometry, Mass Spectrometry & Proteomics, Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Dresden, Germany
| | - Pascal Y Schönberg
- Medical Faculty, University Hospital Carl Gustav Carus, UCC Section Medical Systems Biology, TU Dresden, 01307, Dresden, Germany
| | - Frank Buchholz
- Medical Faculty, University Hospital Carl Gustav Carus, UCC Section Medical Systems Biology, TU Dresden, 01307, Dresden, Germany
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Bernd Schröder
- Institute of Physiological Chemistry, Medizinische Fakultät und Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Torben Mentrup
- Institute of Physiological Chemistry, Medizinische Fakultät und Universitätsklinikum Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
193
|
Son G, Na Y, Kim Y, Son JH, Clemenson GD, Schafer ST, Yoo JY, Parylak SL, Paquola A, Do H, Kim D, Ahn I, Ju M, Kang CS, Ju Y, Jung E, McDonald AH, Park Y, Kim G, Paik SB, Hur J, Kim J, Han YM, Lee SH, Gage FH, Kim JS, Han J. miR-124 coordinates metabolic regulators acting at early stages of human neurogenesis. Commun Biol 2024; 7:1393. [PMID: 39455851 PMCID: PMC11511827 DOI: 10.1038/s42003-024-07089-2] [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: 07/28/2023] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Metabolic dysregulation of neurons is associated with diverse human brain disorders. Metabolic reprogramming occurs during neuronal differentiation, but it is not fully understood which molecules regulate metabolic changes at the early stages of neurogenesis. In this study, we report that miR-124 is a driver of metabolic change at the initiating stage of human neurogenesis. Proteome analysis has shown the oxidative phosphorylation pathway to be the most significantly altered among the differentially expressed proteins (DEPs) in the immature neurons after the knockdown of miR-124. In agreement with these proteomics results, miR-124-depleted neurons display mitochondrial dysfunctions, such as decreased mitochondrial membrane potential and cellular respiration. Moreover, morphological analyses of mitochondria in early differentiated neurons after miR-124 knockdown result in smaller and less mature shapes. Lastly, we show the potential of identified DEPs as novel metabolic regulators in early neuronal development by validating the effects of GSTK1 on cellular respiration. GSTK1, which is upregulated most significantly in miR-124 knockdown neurons, reduces the oxygen consumption rate of neural cells. Collectively, our data highlight the roles of miR-124 in coordinating metabolic maturation at the early stages of neurogenesis and provide insights into potential metabolic regulators associated with human brain disorders characterized by metabolic dysfunctions.
Collapse
Affiliation(s)
- Geurim Son
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Yongwoo Na
- Center for RNA Research, Institute for Basic Science, Seoul, Korea
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Yongsung Kim
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ji-Hoon Son
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Gregory D Clemenson
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Simon T Schafer
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jong-Yeon Yoo
- Department of Biological Sciences, KAIST, Daejeon, Korea
| | - Sarah L Parylak
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Apua Paquola
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Neurology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Hyunsu Do
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Dayeon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Insook Ahn
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Mingyu Ju
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Chanhee S Kang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Younghee Ju
- Department of Biological Sciences, KAIST, Daejeon, Korea
- Sovargen.CO., LTD., Daejeon, Korea
| | - Eunji Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Aidan H McDonald
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Youngjin Park
- Department of Bio and Brain Engineering, KAIST, Daejeon, Korea
| | - Gilhyun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea
| | - Se-Bum Paik
- Department of Bio and Brain Engineering, KAIST, Daejeon, Korea
- Department of Brain and Cognitive Sciences, KAIST, Daejeon, Korea
| | - Junho Hur
- College of Medicine, Hanyang University, Seoul, Korea
| | - Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Yong-Mahn Han
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Seung-Hee Lee
- Department of Biological Sciences, KAIST, Daejeon, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, Korea
- Department of Brain and Cognitive Sciences, KAIST, Daejeon, Korea
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jong-Seo Kim
- Center for RNA Research, Institute for Basic Science, Seoul, Korea
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Jinju Han
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea.
- BioMedical Research Center, KAIST, Daejeon, Korea.
- KAIST Stem Cell Center, KAIST, Daejeon, Korea.
| |
Collapse
|
194
|
Bernardino R, Carvalho AS, Hall MJ, Alves L, Leão R, Sayyid R, Pereira H, Beck HC, Pinheiro LC, Henrique R, Fleshner N, Matthiesen R. Profiling of urinary extracellular vesicle protein signatures from patients with cribriform and intraductal prostate carcinoma in a cross-sectional study. Sci Rep 2024; 14:25065. [PMID: 39443544 PMCID: PMC11500006 DOI: 10.1038/s41598-024-75272-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: 05/11/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
Prognostic tests and treatment approaches for optimized clinical care of prostatic neoplasms are an unmet need. Prostate cancer (PCa) and derived extracellular vesicles (EVs) proteome changes occur during initiation and progression of the disease. PCa tissue proteome has been previously characterized, but screening of tissue samples constitutes an invasive procedure. Consequently, we focused this study on liquid biopsies, such as urine samples. More specifically, urinary small extracellular vesicle and particles proteome profiles of 100 subjects were analyzed using liquid chromatography coupled to high-resolution mass spectrometry (LC-MS/MS). We identified 171 proteins that were differentially expressed between intraductal prostate cancer/cribriform (IDC/Crib) and non-IDC/non-Crib after correction for multiple testing. However, the strong correlation between IDC/Crib and Gleason Grade complicates the disentanglement of the underlying factors driving this association. Nevertheless, even after accounting for multiple testing and adjusting for ISUP (International Society of Urological Pathology) grading, two proteins continued to exhibit significant differential expression between IDC/Crib and non-IDC/non-Crib. Functional enrichment analysis based on cancer hallmark proteins disclosed a clear pattern of androgen response down-regulation in urinary EVs from IDC/Crib compared to non-IDC/non-Crib. Interestingly, proteome differences between IDC and cribriform were more subtle, suggesting high proteome heterogeneity. Overall, the urinary EV proteome reflected partly the prostate pathology.
Collapse
Affiliation(s)
- Rui Bernardino
- Computational and Experimental Biology Group, iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal.
- Division of Urology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada.
| | - Ana Sofia Carvalho
- Computational and Experimental Biology Group, iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal.
| | - Michael J Hall
- Computational and Experimental Biology Group, iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Liliana Alves
- Computational and Experimental Biology Group, iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | | | - Rashid Sayyid
- Division of Urology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Hermínia Pereira
- Department of Pathology, Centro Hospitalar E Universitário Lisboa Central, Lisbon, Portugal
| | - Hans Christian Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry, Odense University Hospital, 5000, Odense, Denmark
| | - Luís Campos Pinheiro
- Department of Urology, Centro Hospitalar e Universitário Lisboa Central, Lisbon, Portugal
| | - Rui Henrique
- Department of Pathology and Cancer Biology and Epigenetics Group - Research Center of IPO Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), R. Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal
- Department of Pathology and Molecular Immunology, ICBAS - School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513, Porto, Portugal
| | - Neil Fleshner
- Division of Urology, Department of Surgical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Rune Matthiesen
- Computational and Experimental Biology Group, iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal.
| |
Collapse
|
195
|
Tiwari M, Gas-Pascual E, Goyal M, Popov M, Matsumoto K, Grafe M, Graf R, Haltiwanger RS, Olszewski N, Orlando R, Samuelson J, West CM. Novel antibodies detect nucleocytoplasmic O-fucose in protist pathogens, cellular slime molds, and plants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618526. [PMID: 39464065 PMCID: PMC11507795 DOI: 10.1101/2024.10.15.618526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Cellular adaptations to change often involve post-translational modifications of nuclear and cytoplasmic proteins. An example found in protists and plants is the modification of serine and threonine residues of dozens to hundreds of nucleocytoplasmic proteins with a single fucose (O-Fuc). A nucleocytoplasmic O-fucosyltransferase (OFT) occurs in the pathogen Toxoplasma gondii, the social amoeba Dictyostelium, and higher plants, where it is called Spy because mutants have a spindly appearance. O-fucosylation, which is required for optimal proliferation of Toxoplasma and Dictyostelium, is paralogous to the O-GlcNAcylation of nucleocytoplasmic proteins of plants and animals that is involved in stress and nutritional responses. O-Fuc was first discovered in Toxoplasma using Aleuria aurantia lectin, but its broad specificity for terminal fucose residues on N- and O-linked glycans in the secretory pathway limits its use. Here we present affinity purified rabbit antisera that are selective for the detection and enrichment of proteins bearing fucose-O-Ser or fucose-O-Thr. These antibodies detect numerous nucleocytoplasmic proteins in Toxoplasma, Dictyostelium, and Arabidopsis, as well as O-Fuc occurring on secretory proteins of Dictyostelium and mammalian cells, although the latter are frequently blocked by further glycosylation. The antibodies label Toxoplasma, Acanthamoeba, and Dictyostelium in a pattern reminiscent of O-GlcNAc in animal cells including nuclear pores. The O-fucome of Dictyostelium is partially conserved with that of Toxoplasma and is highly induced during starvation-induced development. These antisera demonstrate the unique antigenicity of O-Fuc, document conservation of the O-fucome among unrelated protists, and will enable the study of the O-fucomes of other organisms possessing OFT-like genes.
Collapse
Affiliation(s)
- Megna Tiwari
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens GA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens GA
| | - Elisabet Gas-Pascual
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens GA
- Complex Carbohydrate Research Center, University of Georgia, Athens GA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens GA
| | - Manish Goyal
- Department of Molecular and Cell Biology, Boston University School of Medicine, Boston MA
| | | | - Kenjiroo Matsumoto
- Complex Carbohydrate Research Center, University of Georgia, Athens GA
- Current address: Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan; U-Medico Inc., 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Marianne Grafe
- Dept. of Cell Biology, Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
| | - Ralph Graf
- Dept. of Cell Biology, Institute of Biochemistry and Biology, University of Potsdam, 14476 Potsdam, Germany
| | - Robert S. Haltiwanger
- Complex Carbohydrate Research Center, University of Georgia, Athens GA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens GA
| | - Neil Olszewski
- Department of Plant & Microbial Biology, University of Minnesota, 140 Gortner Laboratory, 1479 Gortner Avenue, St. Paul, MN 55108
| | - Ron Orlando
- Complex Carbohydrate Research Center, University of Georgia, Athens GA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens GA
- GlycoScientific LLC, Athens, GA
| | - John Samuelson
- Department of Molecular and Cell Biology, Boston University School of Medicine, Boston MA
| | - Christopher M. West
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens GA
- Complex Carbohydrate Research Center, University of Georgia, Athens GA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens GA
| |
Collapse
|
196
|
Schaefer T, Mittal N, Wang H, Ataman M, Candido S, Lötscher J, Velychko S, Tintignac L, Bock T, Börsch A, Baßler J, Rao TN, Zmajkovic J, Roffeis S, Löliger J, Jacob F, Dumlin A, Schürch C, Schmidt A, Skoda RC, Wymann MP, Hess C, Schöler HR, Zaehres H, Hurt E, Zavolan M, Lengerke C. Nuclear and cytosolic fractions of SOX2 synergize as transcriptional and translational co-regulators of cell fate. Cell Rep 2024; 43:114807. [PMID: 39368083 DOI: 10.1016/j.celrep.2024.114807] [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: 07/14/2023] [Revised: 01/28/2024] [Accepted: 09/13/2024] [Indexed: 10/07/2024] Open
Abstract
Stemness and pluripotency are mediated by transcriptional master regulators that promote self-renewal and repress cell differentiation, among which is the high-mobility group (HMG) box transcription factor SOX2. Dysregulated SOX2 expression, by contrast, leads to transcriptional aberrations relevant to oncogenic transformation, cancer progression, metastasis, therapy resistance, and relapse. Here, we report a post-transcriptional mechanism by which the cytosolic pool of SOX2 contributes to these events in an unsuspected manner. Specifically, a low-complexity region within SOX2's C-terminal segment connects to the ribosome to modulate the expression of cognate downstream factors. Independent of nuclear structures or DNA, this C-terminal functionality alone changes metabolic properties and induces non-adhesive growth when expressed in the cytosol of SOX2 knockout cells. We thus propose a revised model of SOX2 action where nuclear and cytosolic fractions cooperate to impose cell fate decisions via both transcriptional and translational mechanisms.
Collapse
Affiliation(s)
- Thorsten Schaefer
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland.
| | | | - Hui Wang
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Meric Ataman
- Biozentrum, University of Basel, Basel, Switzerland
| | - Silvia Candido
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jonas Lötscher
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Sergiy Velychko
- Max Planck Institute for Molecular Biomedicine, Münster, Germany; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Lionel Tintignac
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Thomas Bock
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Anastasiya Börsch
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jochen Baßler
- Biochemistry Center Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Tata Nageswara Rao
- Medical Research Center, Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland; Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Jakub Zmajkovic
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Sarah Roffeis
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jordan Löliger
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Francis Jacob
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Alain Dumlin
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Christoph Schürch
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Radek C Skoda
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Matthias P Wymann
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Christoph Hess
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; CITIID, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Hans R Schöler
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Holm Zaehres
- Max Planck Institute for Molecular Biomedicine, Münster, Germany; Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Ed Hurt
- Biochemistry Center Heidelberg, Heidelberg University, Heidelberg, Germany
| | | | - Claudia Lengerke
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
197
|
Forero A, Pipicelli F, Moser S, Baumann N, Grätz C, Gonzalez Pisfil M, Pfaffl MW, Pütz B, Kielkowski P, Cernilogar FM, Maccarrone G, Di Giaimo R, Cappello S. Extracellular vesicle-mediated trafficking of molecular cues during human brain development. Cell Rep 2024; 43:114755. [PMID: 39302835 DOI: 10.1016/j.celrep.2024.114755] [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: 11/22/2023] [Revised: 07/03/2024] [Accepted: 08/29/2024] [Indexed: 09/22/2024] Open
Abstract
Cellular crosstalk is an essential process influenced by numerous factors, including secreted vesicles that transfer nucleic acids, lipids, and proteins between cells. Extracellular vesicles (EVs) have been the center of many studies focusing on neurodegenerative disorders, but whether EVs display cell-type-specific features for cellular crosstalk during neurodevelopment is unknown. Here, using human-induced pluripotent stem cell-derived cerebral organoids, neural progenitors, neurons, and astrocytes, we identify heterogeneity in EV protein content and dynamics in a cell-type-specific and time-dependent manner. Our results support the trafficking of key molecules via EVs in neurodevelopment, such as the transcription factor YAP1, and their localization to differing cell compartments depending on the EV recipient cell type. This study sheds new light on the biology of EVs during human brain development.
Collapse
Affiliation(s)
- Andrea Forero
- Max Planck Institute of Psychiatry, Munich, Germany; Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany
| | - Fabrizia Pipicelli
- Max Planck Institute of Psychiatry, Munich, Germany; International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Sylvain Moser
- Max Planck Institute of Psychiatry, Munich, Germany; International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Natalia Baumann
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Christian Grätz
- Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Mariano Gonzalez Pisfil
- Core Facility Bioimaging and Walter-Brendel-Centre of Experimental Medicine, Biomedical Center, Ludwig Maximilian University, Munich, Germany
| | - Michael W Pfaffl
- Division of Animal Physiology and Immunology, Technical University of Munich, Freising, Germany
| | - Benno Pütz
- Max Planck Institute of Psychiatry, Munich, Germany
| | - Pavel Kielkowski
- Department of Chemistry, Ludwig Maximilian University, Munich, Germany
| | - Filippo M Cernilogar
- Division of Molecular Biology, Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University, Munich, Germany; Department of Science and Technological Innovation, University of Piemonte Orientale, Alessandria, Italy
| | | | - Rossella Di Giaimo
- Max Planck Institute of Psychiatry, Munich, Germany; Department of Biology, University of Naples Federico II, Naples, Italy.
| | - Silvia Cappello
- Max Planck Institute of Psychiatry, Munich, Germany; Division of Physiological Genomics, Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University (LMU), Munich, Germany.
| |
Collapse
|
198
|
Hisler V, Bardot P, Detilleux D, Bernardini A, Stierle M, Sanchez EG, Richard C, Arab LH, Ehrhard C, Morlet B, Hadzhiev Y, Jung M, Le Gras S, Négroni L, Müller F, Tora L, Vincent SD. RNA polymerase II transcription initiation in holo-TFIID-depleted mouse embryonic stem cells. Cell Rep 2024; 43:114791. [PMID: 39352809 PMCID: PMC11551524 DOI: 10.1016/j.celrep.2024.114791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 07/09/2024] [Accepted: 09/07/2024] [Indexed: 10/04/2024] Open
Abstract
The recognition of core promoter sequences by TFIID is the first step in RNA polymerase II (Pol II) transcription initiation. Metazoan holo-TFIID is a trilobular complex, composed of the TATA binding protein (TBP) and 13 TBP-associated factors (TAFs). Why and how TAFs are necessary for the formation of TFIID domains and how they contribute to transcription initiation remain unclear. Inducible TAF7 or TAF10 depletion, followed by comprehensive analysis of TFIID subcomplex formation, chromatin binding, and nascent transcription in mouse embryonic stem cells, result in the formation of a TAF7-lacking TFIID or a minimal core-TFIID complex, respectively. These partial complexes support TBP recruitment at promoters and nascent Pol II transcription at most genes early after depletion, but importantly, TAF10 is necessary for efficient Pol II pausing. We show that partially assembled TFIID complexes can sustain Pol II transcription initiation but cannot replace holo-TFIID over several cell divisions and/or development.
Collapse
Affiliation(s)
- Vincent Hisler
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Paul Bardot
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Dylane Detilleux
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Andrea Bernardini
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Matthieu Stierle
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Emmanuel Garcia Sanchez
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Claire Richard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Lynda Hadj Arab
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Cynthia Ehrhard
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Bastien Morlet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France; Proteomics Platform (IGBMC), 67400 Illkirch, France
| | - Yavor Hadzhiev
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Matthieu Jung
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France; GenomEast (IGBMC), 67400 Illkirch, France
| | - Stéphanie Le Gras
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France; GenomEast (IGBMC), 67400 Illkirch, France
| | - Luc Négroni
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France; Proteomics Platform (IGBMC), 67400 Illkirch, France
| | - Ferenc Müller
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - László Tora
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France
| | - Stéphane D Vincent
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67400 Illkirch, France; CNRS, UMR7104, 67400 Illkirch, France; INSERM, U1258, 67400 Illkirch, France; Université de Strasbourg, 67400 Illkirch, France.
| |
Collapse
|
199
|
Grenier C, Lin IH, Peters D, Pozzi A, Lennon R, Naylor RW. Integrin alpha1 beta1 promotes interstitial fibrosis in a mouse model of polycystic kidney disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619080. [PMID: 39484448 PMCID: PMC11526950 DOI: 10.1101/2024.10.18.619080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Fibrosis is the cause of end-stage kidney failure in patients with Autosomal Dominant Polycystic Kidney Disease (ADPKD). The molecular and cellular mechanisms involved in fibrosis are complex and anti-fibrotic therapies have so far failed to make an impact on patient welfare. Using unbiased proteomics analysis on the Pkd1 nl/nl mouse, we found that expression of the integrin α1 subunit is increased in this model of ADPKD. In human ADPKD tissue and two single cell RNA kidney disease datasets, ITGA1 was also upregulated. To investigate the functional role of this integrin subunit in ADPKD, we generated a Pkd1 nl/nl Itga1 -/- mouse. We observed a significant reduction in kidney volume and kidney dysfunction in mice lacking the integrin α1 subunit. Kidneys from Pkd1 nl/nl Itga1 -/- mice had smaller cysts and reduced interstitial expansion and tubular atrophy. Picrosirius red staining identified a restriction in collagen staining in the interstitium and the myofibroblast marker α smooth muscle actin was also downregulated. Myofibroblast cell proliferation was reduced in Pkd1 nl/nl Itga1 -/- mice and primary fibroblast cultures demonstrated an abrogated fibrogenic phenotype in integrin α1-depleted fibroblasts. These results highlight a previously unrecognised role for the integrin α1 subunit in kidney fibrosis.
Collapse
Affiliation(s)
- C Grenier
- Manchester Cell-Matrix Centre, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - I-H Lin
- Bioinformatics Core Facility, Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Djm Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - A Pozzi
- Department of Medicine, Division of Nephrology and Hypertension
- Department of Veterans Affairs, Nashville, Tennessee, USA
| | - R Lennon
- Manchester Cell-Matrix Centre, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - R W Naylor
- Manchester Cell-Matrix Centre, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
200
|
Wen B, Hsu C, Zeng WF, Riffle M, Chang A, Mudge M, Nunn B, Berg MD, Villén J, MacCoss MJ, Noble WS. Carafe enables high quality in silico spectral library generation for data-independent acquisition proteomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618504. [PMID: 39463980 PMCID: PMC11507862 DOI: 10.1101/2024.10.15.618504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Data-independent acquisition (DIA)-based mass spectrometry is becoming an increasingly popular mass spectrometry acquisition strategy for carrying out quantitative proteomics experiments. Most of the popular DIA search engines make use of in silico generated spectral libraries. However, the generation of high-quality spectral libraries for DIA data analysis remains a challenge, particularly because most such libraries are generated directly from data-dependent acquisition (DDA) data or are from in silico prediction using models trained on DDA data. In this study, we developed Carafe, a tool that generates high-quality experiment-specific in silico spectral libraries by training deep learning models directly on DIA data. We demonstrate the performance of Carafe on a wide range of DIA datasets, where we observe improved fragment ion intensity prediction and peptide detection relative to existing pretrained DDA models.
Collapse
Affiliation(s)
- Bo Wen
- Department of Genome Sciences, University of Washington
| | - Chris Hsu
- Department of Genome Sciences, University of Washington
| | - Wen-Feng Zeng
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Germany
| | | | - Alexis Chang
- Department of Genome Sciences, University of Washington
| | - Miranda Mudge
- Department of Genome Sciences, University of Washington
| | - Brook Nunn
- Department of Genome Sciences, University of Washington
| | | | - Judit Villén
- Department of Genome Sciences, University of Washington
| | | | - William S. Noble
- Department of Genome Sciences, University of Washington
- Paul G. Allen School of Computer Science and Engineering, University of Washington
| |
Collapse
|