1
|
Kamble NS, Thomas S, Madaan T, Ehsani N, Sange S, Tucker K, Muhumure A, Kunkler S, Kotagiri N. Engineered bacteria as an orally administered anti-viral treatment and immunization system. Gut Microbes 2025; 17:2500056. [PMID: 40340796 PMCID: PMC12064065 DOI: 10.1080/19490976.2025.2500056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/05/2025] [Accepted: 04/24/2025] [Indexed: 05/10/2025] Open
Abstract
The emergence of new viral pathogens necessitates innovative antiviral therapies and vaccines. Traditional approaches, such as monoclonal antibodies and vaccines, are often hindered by resistance, limited effectiveness, and high costs. Here, we develop an engineered probiotic-based antiviral platform using Escherichia coli Nissle 1917 (EcN), capable of providing both mucosal and systemic immunity via oral administration. EcN was engineered to display anti-spike nanobodies or express the Spike-Receptor Binding Domain on its surface. Our findings reveal that EcN with nanobodies effectively inhibits the interaction between spike protein-expressing pseudoviruses and the ACE2 receptor. Furthermore, we observed the translocation of nanobodies to distant organs, facilitated by outer membrane vesicles (OMVs). The oral administration of EcN expressing spike proteins induced a robust immune response characterized by the production of both IgG and IgA, antibodies that blocked the pseudovirus-ACE2 interaction. While SARS-CoV-2 served as a model, this versatile probiotic platform holds potential for developing customizable biotherapeutics against a wide range of emerging pathogens such as influenza virus or respiratory syncytial virus (RSV) by engineering EcN to express viral surface protein or neutralizing nanobodies demonstrating its versatility as a next-generation mucosal vaccine strategy.
Collapse
Affiliation(s)
- Nitin S. Kamble
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Shindu Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Tushar Madaan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Nadia Ehsani
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Saqib Sange
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Kiersten Tucker
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Alexis Muhumure
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Sarah Kunkler
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
2
|
Fuchs S, Fiedler MK, Heiduk N, Wanisch A, Mibus C, Singh D, Debowski AW, Marshall BJ, Vieth M, Josenhans C, Suerbaum S, Sieber SA, Gerhard M, Mejías-Luque R. Helicobacter pylori γ-glutamyltransferase is linked to proteomic adaptions important for colonization. Gut Microbes 2025; 17:2488048. [PMID: 40205659 PMCID: PMC11988274 DOI: 10.1080/19490976.2025.2488048] [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: 08/26/2024] [Revised: 03/18/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Helicobacter pylori γ-glutamyltransferase (gGT) is a virulence factor that promotes bacterial colonization and immune tolerance. Although some studies addressed potential functional mechanisms, the supportive role of gGT for in vivo colonization remains unclear. Additionally, it is unknown how different gGT expression levels may lead to compensatory mechanisms ensuring infection and persistence. Hence, it is crucial to unravel the in vivo function of gGT. We assessed acid survival under conditions mimicking the human gastric fluid and elevated the pH in the murine stomach prior to H. pylori infection to link gGT-mediated acid resistance to colonization. By comparing proteomes of gGT-proficient and -deficient isolates before and after infecting mice, we investigated proteomic adaptations of gGT-deficient bacteria during infection. Our data indicate that gGT is crucial to sustain urease activity in acidic environments, thereby supporting survival and successful colonization. Absence of gGT triggers expression of proteins involved in the nitrogen and iron metabolism and boosts the expression of adhesins and flagellar proteins during infection, resulting in increased motility and adhesion capacity. In summary, gGT-dependent mechanisms confer a growth advantage to the bacterium in the gastric environment, which renders gGT a valuable target for the development of new treatments against H. pylori infection.
Collapse
Affiliation(s)
- Sonja Fuchs
- Institute for Medical Microbiology, Immunology and Hygiene, Department of Preclinical Medicine, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Michaela K. Fiedler
- Center for Functional Protein Assemblies (CPA), Chair of Organic Chemistry II, Department Biosciences, TUM School of Natural Sciences, Technical University of Munich (TUM), Garching, Germany
| | - Nicole Heiduk
- Institute for Medical Microbiology, Immunology and Hygiene, Department of Preclinical Medicine, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Andreas Wanisch
- Institute for Medical Microbiology, Immunology and Hygiene, Department of Preclinical Medicine, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Cora Mibus
- Institute for Medical Microbiology, Immunology and Hygiene, Department of Preclinical Medicine, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Dharmesh Singh
- Institute for Medical Microbiology, Immunology and Hygiene, Department of Preclinical Medicine, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Aleksandra W. Debowski
- Marshall Centre for Infectious Disease Research and Training, School of Biomedical Sciences, The University of Western Australia, Nedlands, Australia
- School of Molecular Sciences, The University of Western Australia, Crawley, Australia
| | - Barry J. Marshall
- Marshall Centre for Infectious Disease Research and Training, School of Biomedical Sciences, The University of Western Australia, Nedlands, Australia
| | - Michael Vieth
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nuremberg, Klinikum Bayreuth, Bayreuth, Germany
| | - Christine Josenhans
- Max von Pettenkofer Institute, Faculty of Medicine, Medical Microbiology and Hospital Epidemiology, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- DZIF - German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Sebastian Suerbaum
- Max von Pettenkofer Institute, Faculty of Medicine, Medical Microbiology and Hospital Epidemiology, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- DZIF - German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Stephan A. Sieber
- Center for Functional Protein Assemblies (CPA), Chair of Organic Chemistry II, Department Biosciences, TUM School of Natural Sciences, Technical University of Munich (TUM), Garching, Germany
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, Department of Preclinical Medicine, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, Department of Preclinical Medicine, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| |
Collapse
|
3
|
Rommelaere S, Schüpfer F, Armand F, Hamelin R, Lemaitre B. An updated proteomic analysis of Drosophila haemolymph after bacterial infection. Fly (Austin) 2025; 19:2485685. [PMID: 40223358 PMCID: PMC12005426 DOI: 10.1080/19336934.2025.2485685] [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: 12/18/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
Using an in-depth Mass Spectrometry-based proteomics approach, we provide a comprehensive characterization of the hemolymphatic proteome of adult flies upon bacterial infection. We detected and quantified changes in abundance of several known immune regulators and effectors, including multiple antimicrobial peptides, peptidoglycan-binding proteins and serine proteases. Comparison to previously published transcriptomic analyses reveals a partial overlap with our dataset, indicating that many proteins released into the haemolymph upon infection may not be regulated at the transcript level. Among them, we identify a set of muscle-derived proteins released into the haemolymph upon infection. Finally, our analysis reveals that infection induces major changes in the abundance of proteins associated with mitochondrial respiration. This study uncovers a large number of previously undescribed proteins potentially involved in the immune response.
Collapse
Affiliation(s)
- Samuel Rommelaere
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Fanny Schüpfer
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Florence Armand
- EPFL Proteomics Core Facility, EPFL SV PTECH PTP, Lausanne, Switzerland
| | - Romain Hamelin
- EPFL Proteomics Core Facility, EPFL SV PTECH PTP, Lausanne, Switzerland
| | - Bruno Lemaitre
- Global Health Institute, School of Life Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
4
|
Purohit K, Pathak R, Hayes E, Sunna A. Novel bioactive peptides from ginger rhizome: Integrating in silico and in vitro analysis with mechanistic insights through molecular docking. Food Chem 2025; 484:144432. [PMID: 40279907 DOI: 10.1016/j.foodchem.2025.144432] [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: 11/26/2024] [Revised: 04/14/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Ginger (Zingiber officinale) is widely recognised for its functional benefits, primarily attributed to its diverse phytochemicals. However, its proteome remains largely unexplored. This study hypothesised that isolated peptides may exhibit different bioactivities or more targeted mechanisms of action and could be investigated at a molecular level. Proteins were enzymatically hydrolysed under five conditions, and peptides were identified using LC-MS/MS. In silico screening suggested antioxidant, ACE-inhibitory, and antibacterial properties, further assessed through molecular docking and in vitro validation. 41 potentially bioactive peptides were identified. In vitro assays confirmed these properties for selected peptides, P1 (GSPVWIIPEPT), P2 (FASYPVKK), P3 (GPEKIFYDGPYL), and P4 (IAISPSYPIK). Notably, P4 exhibited potent mixed-type ACE-inhibition and bacteriostatic effects. Molecular docking provided mechanistic insights into these interactions. These findings highlight ginger as a promising source of bioactive peptides while underscoring the need to complement AI tools with in vitro and in vivo validations due to observed discrepancies.
Collapse
Affiliation(s)
- Kruttika Purohit
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia; Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia's Bioactives (FAAB), Sydney, NSW 2109, Australia
| | - Rachana Pathak
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia; Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia's Bioactives (FAAB), Sydney, NSW 2109, Australia
| | - Evan Hayes
- Factors Group Australia, Sydney, NSW 2116, Australia
| | - Anwar Sunna
- School of Natural Sciences, Macquarie University, Sydney, NSW 2109, Australia; Australian Research Council Industrial Transformation Training Centre for Facilitated Advancement of Australia's Bioactives (FAAB), Sydney, NSW 2109, Australia.
| |
Collapse
|
5
|
Richter P, Karanth S, Dos Santos Natividade R, Nicoli A, Kogut-Guenthel MM, Benthin J, Di Pizio A, Koehler M, Somoza V. Biomolecular and biophysical AFM probing reveals distinct binding of bitter peptide VAPFPEVF to TAS2R16 without inducing an intracellular calcium response. Food Chem 2025; 484:144448. [PMID: 40288211 DOI: 10.1016/j.foodchem.2025.144448] [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: 12/17/2024] [Revised: 03/17/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
The casein-derived bitter peptide VAPFPEVF has been shown to stimulate proton secretion in human parietal cells (HGT-1) via bitter taste receptor TAS2R16, confirmed by siRNA knockdown. Since literature evidence is inconclusive, we hypothized that VAPFPEVF binds to TAS2R16, and investigated its effects on G protein-coupled signaling pathways. Exposure of HGT-1 cells to VAPFPEVF altered cAMP signaling without inducing a calcium response. An atomic force microscopy (AFM)-based approach was employed to demonstrate peptide binding to TAS2R16 in cellular and cell-free environments using TAS2R16-reconstituted proteoliposomes. Increased binding events were observed, reduced by the addition of salicin and TAS2R16 antagonist probenecid. AlphaFold multimer and molecular dynamics simulations suggest VAPFPEVF binds the orthosteric site of TAS2R16. These findings reveal (i) VAPFPEVF interacts with TAS2R16 to modulate cAMP levels without triggering calcium mobilization and (ii) the AFM approach as a valuable tool for studying peptide binding to TAS2R16 and possibly other G-protein coupled transmembrane receptors.
Collapse
Affiliation(s)
- Phil Richter
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354 Freising, Germany; Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany.
| | - Sanjai Karanth
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany.
| | - Rita Dos Santos Natividade
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany.
| | - Alessandro Nicoli
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354 Freising, Germany; Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany.
| | - Małgorzata M Kogut-Guenthel
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany.
| | - Julia Benthin
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354 Freising, Germany; Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany.
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany; Chemoinformatics and Protein Modelling, TUM School of Life Sciences, Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany.
| | - Melanie Koehler
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany; TUM Junior Fellow at the Chair of Nutritional Systems Biology at the Technical University of Munich, 85354, Freising, Germany.
| | - Veronika Somoza
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany; Chair of Nutritional Systems Biology, TUM School of Life Sciences, Technical University of Munich, Lise-Meitner-Str. 34, 85354 Freising, Germany; Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), 1090 Wien, Austria.
| |
Collapse
|
6
|
Duft RG, Griffin JL, Stead DA. MEATiCode: A comprehensive proteomic LC-MS/MS method for simultaneous species identification in meat authentication. Food Chem 2025; 483:144231. [PMID: 40220447 DOI: 10.1016/j.foodchem.2025.144231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/10/2024] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
Food fraud in the meat industry threatens consumer trust, market stability, and public health. Traditional methods like DNA barcoding are limited, especially for processed foods where DNA is often degraded. This paper introduces the workflow MEATiCode, a comprehensive proteomic liquid chromatography tandem mass spectrometry (LC-MS/MS) method for the simultaneous identification of species in meat authentication. Application of a novel database search approach - MEATiCode - enabled the differentiation of meat species (as demonstrated for beef, pork, chicken and lamb) in raw and cooked food products following a simple sample preparation procedure and LC-MS/MS analysis of extracted meat peptides. The efficacy of the MEATiCode method was demonstrated through its application to a range of meat products, achieving high sensitivity (0.5 % Limit of Detection (LoD)) and reliability in the detection of adulteration, even in highly processed or cooked meats.
Collapse
Affiliation(s)
- Renata G Duft
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Ashgrove Rd W, Aberdeen AB25 2ZD, UK.
| | - Julian L Griffin
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Ashgrove Rd W, Aberdeen AB25 2ZD, UK
| | - David A Stead
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Ashgrove Rd W, Aberdeen AB25 2ZD, UK
| |
Collapse
|
7
|
Cherkaoui M, Le Corre E, Ahmat-Sougoudi A, Perrin E, Solé-Jamault V, Rabesona H, Denery-Papini S, Morisset M, Rogniaux H, Dijk W. Exploring the molecular modifications and allergenicity of the egg white protein matrix during boiling. Food Chem 2025; 483:144304. [PMID: 40239576 DOI: 10.1016/j.foodchem.2025.144304] [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: 12/18/2024] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Hen's egg allergy is the second most common food allergy in young children, with the major allergens ovalbumin and ovomucoid found in egg white. While many egg-allergic children can tolerate baked or hard-boiled eggs, there is limited understanding of how heating affects allergen structure and allergenicity within the egg white protein matrix. This study investigated the impact of egg white boiling for 10 or 45 min on the structure and allergenicity of the main egg white allergens. Our results showed that 45 min of boiling led to significant structural changes and a strong reduction in egg white allergenicity, while 10 min of boiling had a minor effect. Gastric digestion further reduced allergenicity, especially in 45 min boiled eggs. These findings highlight how increasing the boiling time of egg white can reduce allergenicity through structural changes in the main egg white allergens.
Collapse
Affiliation(s)
- Mehdi Cherkaoui
- INRAE, UR1268 BIA, F-44316, Nantes, France; INRAE, PROBE Research Infrastructure, BIBS Facility, F-44316, Nantes, France
| | | | | | | | | | | | | | | | - Hélène Rogniaux
- INRAE, UR1268 BIA, F-44316, Nantes, France; INRAE, PROBE Research Infrastructure, BIBS Facility, F-44316, Nantes, France
| | | |
Collapse
|
8
|
Johansson Å, Venkita Subramani M, Yilmaz B, Nyström EE, Layunta E, Arike L, Sommer F, Rosenstiel P, Vereecke L, Mannerås-Holm L, Wullaert A, Pelaseyed T, Johansson ME, Birchenough GM. Neonatal microbiota colonization primes maturation of goblet cell-mediated protection in the pre-weaning colon. J Exp Med 2025; 222:e20241591. [PMID: 40323318 PMCID: PMC12051479 DOI: 10.1084/jem.20241591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 02/06/2025] [Accepted: 04/03/2025] [Indexed: 05/08/2025] Open
Abstract
Regulated host-microbe interactions are a critical aspect of lifelong health. Colonic goblet cells protect from microorganisms via the generation of a mucus barrier structure. Bacteria-sensing sentinel goblet cells provide secondary protection by orchestrating mucus secretion when microbes breach the mucus barrier. Mucus deficiencies in germ-free mice implicate a role for the microbiota in programming barrier generation, but its natural ontogeny remains undefined. We now investigate the mucus barrier and sentinel goblet cell development in relation to postnatal colonization. Combined in vivo and ex vivo analyses demonstrate rapid and sequential microbiota-dependent development of these primary and secondary goblet cell protective functions, with dynamic changes in mucus processing dependent on innate immune signaling via MyD88 and development of functional sentinel goblet cells dependent on the NADPH/dual oxidase family member Duox2. Our findings identify new mechanisms of microbiota-goblet cell regulatory interaction and highlight the critical importance of the pre-weaning period for the normal development of protective systems that are key legislators of host-microbiota interaction.
Collapse
Affiliation(s)
- Åsa Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Mahadevan Venkita Subramani
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Bahtiyar Yilmaz
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Elisabeth E.L. Nyström
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Elena Layunta
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Liisa Arike
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Felix Sommer
- Institute of Clinical & Molecular Biology, University of Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical & Molecular Biology, University of Kiel, Kiel, Germany
| | - Lars Vereecke
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Louise Mannerås-Holm
- Wallenberg Laboratory, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Andy Wullaert
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Biomedical Sciences, Cell Death Signalling Lab, University of Antwerp, Antwerp, Belgium
| | - Thaher Pelaseyed
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Malin E.V. Johansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - George M.H. Birchenough
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
9
|
Monteiro R, Alcantud BS, Piersma S, Hendrickx APA, Maaß S, Becher D, Azeredo J, Bathoorn E, van Dijl JM. Outer membrane vesicles of carbapenem-resistant clinical Acinetobacter baumannii isolates protect both the vesicle-producing bacteria and non-resistant bacteria against carbapenems. Microbiol Res 2025; 297:128175. [PMID: 40239429 DOI: 10.1016/j.micres.2025.128175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 01/23/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025]
Abstract
Infections caused by carbapenem-resistant Acinetobacter baumannii (A. baumannii; CRAb) are associated with high patient morbidity and mortality. The serious threat for human health imposed by CRAb was recently underscored by identification of close-to-untouchable carbapenem- and tetracycline-resistant isolates. Since outer membrane vesicles (OMVs) of Gram-negative bacteria may contribute to antimicrobial resistance, our present study was aimed at investigating OMVs produced by the first two carbapenem- and tetracycline-resistant A. baumannii isolates in Europe. These isolates, denoted CRAb1 and CRAb2, contain large, nearly identical plasmids that specify multiple resistances. Both isolates produce OMVs that were analyzed by differential light scattering, transmission electron microscopy and proteomics. By comparison with OMVs from the plasmid-free non-carbapenem-resistant A. baumannii isolate Ab1, which is an isogenic ancestor of the CRAb1 isolate, we show that plasmid carriage by the CRAb1 and CRAb2 isolates leads to an increased OMV size that is accompanied by increased diversity of the OMV proteome. Our analyses show that OMVs from CRAb1 and CRAb2 are major reservoirs of proteins involved in antimicrobial resistance, including the plasmid-encoded carbapenemases New Delhi metallo-β-lactamase-1 (NDM-1), and carbapenem-hydrolyzing oxacillinase OXA-97 (OXA-97). Here we report that these OMV-borne carbapenemases hydrolyze imipenem and protect otherwise carbapenem-sensitive A. baumannii and Escherichia coli (E. coli) isolates against this antibiotic. In conclusion, our findings demonstrate that OMVs from highly drug-resistant CRAb confer protection against last-resort antibiotics to non-resistant bacterial pathogens.
Collapse
Affiliation(s)
- Rodrigo Monteiro
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands; Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Beatriz Santamarina Alcantud
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands
| | - Sjouke Piersma
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands
| | - Antoni P A Hendrickx
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Sandra Maaß
- University of Greifswald, Centre of Functional Genomics of Microbes, Department of Microbial Proteomics, Institute of Microbiology, Greifswald, Germany
| | - Dörte Becher
- University of Greifswald, Centre of Functional Genomics of Microbes, Department of Microbial Proteomics, Institute of Microbiology, Greifswald, Germany
| | - Joana Azeredo
- Centre of Biological Engineering, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Erik Bathoorn
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands
| | - Jan Maarten van Dijl
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands.
| |
Collapse
|
10
|
Caliendo A, Camorani S, Ibarra LE, Pinto G, Agnello L, Albanese S, Caianiello A, Illiano A, Festa R, Ambrosio V, Scognamiglio G, Cantile M, Amoresano A, Fedele M, Zannetti A, Cerchia L. A novel CD44-targeting aptamer recognizes chemoresistant mesenchymal stem-like TNBC cells and inhibits tumor growth. Bioact Mater 2025; 50:443-460. [PMID: 40342488 PMCID: PMC12059597 DOI: 10.1016/j.bioactmat.2025.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/20/2025] [Indexed: 05/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) represents a significant therapeutic challenge owing to the scarcity of targeted medicines and elevated recurrence rates. We previously reported the development of the nuclease-resistant RNA sTN58 aptamer, which selectively targets TNBC cells. Here, sTN58 aptamer was employed to capture and purify its binding target from the membrane protein fraction of cisplatin-resistant mesenchymal stem-like TNBC cells. Mass spectrometry in conjunction with aptamer binding assays across various cancer cell lines identified CD44 as the cellular target of sTN58. By binding to CD44, sTN58 inhibits the invasive growth and hyaluronic acid-dependent tube formation in chemoresistant TNBC cells, where CD44 serves as a key driver of tumor cell aggressiveness and stem-like plasticity. Moreover, in vivo studies demonstrated the aptamer's high tumor targeting efficacy and its capacity to significantly inhibit tumor growth and lung metastases following intravenous administration in mice with orthotopic TNBC. Overall, our findings reveal the striking potential of sTN58 as a targeting reagent for the recognition and therapy of cancers overexpressing CD44.
Collapse
Affiliation(s)
- Alessandra Caliendo
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Simona Camorani
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Luis Exequiel Ibarra
- Institute of Environmental Biotechnology and Health (INBIAS), National University of Rio Cuarto (UNRC), National Council for Scientific and Technological Research (CONICET), Río Cuarto, X5800BIA, Argentina
| | - Gabriella Pinto
- Dipartimento di Scienze Chimiche Università di Napoli Federico II, Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Lisa Agnello
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Sandra Albanese
- Institute of Biostructures and Bioimaging, National Research Council, 80145, Naples, Italy
| | - Antonietta Caianiello
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Anna Illiano
- Dipartimento di Scienze Chimiche Università di Napoli Federico II, Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Rosaria Festa
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Vincenzo Ambrosio
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Giosuè Scognamiglio
- Institutional Biobank-Scientific Directorate, National Cancer Institute INT-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Monica Cantile
- Institutional Biobank-Scientific Directorate, National Cancer Institute INT-IRCCS Fondazione G. Pascale, 80131, Naples, Italy
| | - Angela Amoresano
- Dipartimento di Scienze Chimiche Università di Napoli Federico II, Consorzio Interuniversitario Istituto Nazionale Biostrutture e Biosistemi, Roma, Italy
| | - Monica Fedele
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging, National Research Council, 80145, Naples, Italy
| | - Laura Cerchia
- Institute of Endotypes in Oncology, Metabolism and Immunology "Gaetano Salvatore", National Research Council, 80131, Naples, Italy
| |
Collapse
|
11
|
Gradl K, Richter P, Somoza V. Bitter peptides formed during in-vitro gastric digestion induce mechanisms of gastric acid secretion and release satiating serotonin via bitter taste receptors TAS2R4 and TAS2R43 in human parietal cells in culture. Food Chem 2025; 482:144174. [PMID: 40184744 DOI: 10.1016/j.foodchem.2025.144174] [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: 10/02/2024] [Revised: 03/02/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
A key barrier in transitioning to plant-based, more satiating diets, is the bitter taste of plant proteins. We hypothesize that both, a more bitter tasting (MBT) and a less bitter tasting (LBT) pea protein hydrolysate (PPH) can be digested in the stomach into bitter tasting peptides that stimulate proton secretion (PS) and serotonin release, as two of the key gastric satiety signals, via the functional involvement of bitter taste receptors (TAS2Rs). Using a sensory-guided LC-MS approach, we identified six bitter peptides that were released from LBT-PPH and MBT-PPH during gastric digestion in vitro. TAS2R4 and TAS2R43 involvement in PS and serotonin release was confirmed via CRISPR-Cas9 knockout experiments. Our hypothesis was proven with all six peptides equally stimulating PS in immortalized human gastric HGT-1 cells, and LBT-PPH-derived peptides eliciting a higher serotonin release in HGT-1 cells than MBT-PPH peptides, indicating a satiating potential of less bitter tasting protein hydrolysates.
Collapse
Affiliation(s)
- Katrin Gradl
- TUM School of Life Sciences, Technical University of Munich, Alte Akademie 8, 85354 Freising, Germany; Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany
| | - Phil Richter
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany
| | - Veronika Somoza
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany; Chair of Nutritional Systems Biology, Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany; Department of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| |
Collapse
|
12
|
Xu J, CailianWang, Liu T, Luo R, Zheng C, Zhang Y, Lang X. Meat quality differences and protein molecular mechanisms affecting meat flavor in different breeds of Tibetan sheep analyzed by 4D label-free quantitative proteomics. Food Chem 2025; 480:143977. [PMID: 40138833 DOI: 10.1016/j.foodchem.2025.143977] [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: 12/17/2024] [Revised: 02/22/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025]
Abstract
To evaluate the meat quality of the new breed of Panou sheep, the longissimus dorsi (LD) muscles of 1.5-year-old Panou sheep and the local breed of Oula sheep were selected for comparative analysis in terms of meat quality, and the molecular mechanisms influencing flavor were investigated using 4D label-free proteomics technology. The results revealed that the fiber density, tenderness, and brightness of the Panou sheep meat were lower than those of the Oula sheep, and the composition of amino acids and flavor substances made it possible to determine that the Panou sheep meat has a high-quality and distinctive flavor. Proteomic analysis indicated that the metabolic pathways that may be associated with meat flavor are amino acid catabolism and sugar metabolism. This study explored the role of proteins in the regulation of meat flavor in Tibetan sheep, which provides a reference for the identification of meat products and subsequent breed improvement.
Collapse
Affiliation(s)
- Jianfeng Xu
- Institute of Animal & Pasture Science and Green Agriculture, Gansu Academy of Agricultural Science, Lanzhou 730070, China; College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - CailianWang
- Institute of Animal & Pasture Science and Green Agriculture, Gansu Academy of Agricultural Science, Lanzhou 730070, China
| | - Ting Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Ruirui Luo
- Institute of Animal & Pasture Science and Green Agriculture, Gansu Academy of Agricultural Science, Lanzhou 730070, China
| | - Chen Zheng
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Yanshu Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Xia Lang
- Institute of Animal & Pasture Science and Green Agriculture, Gansu Academy of Agricultural Science, Lanzhou 730070, China.
| |
Collapse
|
13
|
Ferreira JV, Ahmed Y, Heunis T, Jain A, Johnson E, Räschle M, Ernst R, Vanni S, Carvalho P. Pex30-dependent membrane contact sites maintain ER lipid homeostasis. J Cell Biol 2025; 224:e202409039. [PMID: 40407417 PMCID: PMC12101078 DOI: 10.1083/jcb.202409039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/28/2025] [Accepted: 03/12/2025] [Indexed: 05/26/2025] Open
Abstract
In eukaryotic cells, communication between organelles and the coordination of their activities depend on membrane contact sites (MCS). How MCS are regulated under the dynamic cellular environment remains poorly understood. Here, we investigate how Pex30, a membrane protein localized to the endoplasmic reticulum (ER), regulates multiple MCS in budding yeast. We show that Pex30 is critical for the integrity of ER MCS with peroxisomes and vacuoles. This requires the dysferlin (DysF) domain on the Pex30 cytosolic tail. This domain binds to phosphatidic acid (PA) both in vitro and in silico, and it is important for normal PA metabolism in vivo. The DysF domain is evolutionarily conserved and may play a general role in PA homeostasis across eukaryotes. We further show that the ER-vacuole MCS requires a Pex30 C-terminal domain of unknown function and that its activity is controlled by phosphorylation in response to metabolic cues. These findings provide new insights into the dynamic nature of MCS and their coordination with cellular metabolism.
Collapse
Affiliation(s)
| | - Yara Ahmed
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Tiaan Heunis
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Aamna Jain
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
- Preclinical Center for Molecular Signaling, Saarland University, Homburg, Germany
| | - Errin Johnson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Markus Räschle
- Department of Molecular Genetics, TU Kaiserslautern, Kaiserslautern, Germany
| | - Robert Ernst
- Medical Biochemistry and Molecular Biology, Saarland University, Homburg, Germany
- Preclinical Center for Molecular Signaling, Saarland University, Homburg, Germany
| | - Stefano Vanni
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Swiss National Center for Competence in Research Bio-inspired Materials, University of Fribourg, Fribourg, Switzerland
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| |
Collapse
|
14
|
von Ehr A, Steenbuck ID, Häfele C, Remmersmann F, Vico TA, Ehlert C, Lindner D, Wolf D, Tholen S, Schilling O, Czerny M, Westermann D, Hilgendorf I. Experimental evidence on colchicine's mode of action in human carotid artery plaques. Atherosclerosis 2025; 406:119239. [PMID: 40381496 DOI: 10.1016/j.atherosclerosis.2025.119239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/27/2025] [Accepted: 05/03/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND AND AIMS Atherosclerosis, driven by inflammation, is a leading cause of cardiovascular events. Recent clinical trials have highlighted the therapeutic potential of anti-inflammatory treatments. Consequently, colchicine is being recommended for secondary prevention in current guidelines, although the drug's mechanistic actions are not fully understood. METHODS To this end, we conducted a multiomic investigation of colchicine's effect on human carotid plaques. Sections from endarterectomy specimens were exposed to colchicine at concentrations of 2 ng/ml and 10 ng/ml ex vivo for 24 h and compared to untreated segments of the same plaque. Gene expression changes were analyzed by bulk RNA sequencing, and plaque secretomes underwent mass spectrometry for proteomic analysis. In situ cell proliferation was assessed by histology. RESULTS Our data indicate, that colchicine suppresses neutrophil and platelet degranulation and activation, collagen degradation and atheromatous plaque macrophage proliferation in a dose-dependent manner in human plaques, while stimulating myofibroblast activation. Unexpectedly, interleukine (IL)-1beta release from colchicine treated plaques was not reduced. These results indicate that the inflammasome may not be the predominant target of low-dose colchicine in human carotid artery plaques. CONCLUSION Our study identifies multifactorial pathways through which colchicine, the first cardiovascular guideline-recommended anti-inflammatory drug, predominantly acts on human atherosclerotic lesions beyond the inflammasome. Targeting neutrophil and platelet degranulation, collagen degradation and macrophage proliferation, selectively, may provide substantial therapeutic benefit in atherosclerotic cardiovascular disease without colchicine's undesired side effects.
Collapse
Affiliation(s)
- Alexander von Ehr
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Ines Derya Steenbuck
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Charlotte Häfele
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felix Remmersmann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tamara A Vico
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carolin Ehlert
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Tholen
- Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Institute for Surgical Pathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Martin Czerny
- Department of Cardiovascular Surgery, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Institute of Experimental Cardiovascular Medicine, University Heart Center Freiburg-Bad Krozingen, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
15
|
Roncero E, Álvarez M, Cerrada L, Delgado J, Andrade MJ. Debaryomyces hansenii alone and in combination with plant extracts reduce ochratoxin A in dry-cured "chorizo". Food Res Int 2025; 212:116512. [PMID: 40382059 DOI: 10.1016/j.foodres.2025.116512] [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: 12/23/2024] [Revised: 04/13/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025]
Abstract
The presence of ochratoxin A (OTA) in dry-cured sausages is a current hazard. To control its contamination, the use of biocontrol agents (BCAs) of plant and microbial origin as anti-ochratoxigenic strategies is being carried out. The aim of this study was to evaluate the anti-ochratoxigenic effect of rosemary essential oil (REO), acorn shell extract (AE) and Debaryomyces hansenii against OTA production by Penicillium nordicum in the Spanish dry-cured sausages "chorizo". For this purpose, BCAs were individually and in combination inoculated in the presence of P. nordicum on the surface of portions of "chorizo" and incubated under typical ripening conditions. Samples were taken for analysing OTA production and proteomic profile variation as well as for physico-chemical and sensory analyses of the sausage portions. Individual application of REO and AE significantly increased OTA production, likely as a response to stressful stimuli. On the other hand, treatments that included D. hansenii significantly decreased it, apparently due to the influence of this yeast on triggering OTA degradation processes of P. nordicum. Although physico-chemical and sensory characteristics were altered in some cases, the degree of acceptability was high. These results reflect the possibility of using D. hansenii as an effective BCA with the ability to mitigate increased OTA production in the presence of plant-derived BCAs inoculated on the surface of "chorizo". Additionally, the sensory results suggest a plausible industrial application due to the absence of negative effects on the final product.
Collapse
Affiliation(s)
- Elia Roncero
- Higiene y Seguridad Alimentaria, Instituto Universitario de Investigación de Carne y Productos Cárnicos, Facultad de Veterinaria, Universidad de Extremadura, 10003 Cáceres, Spain
| | - Micaela Álvarez
- Sección Departamental de Nutrición y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Lucía Cerrada
- Higiene y Seguridad Alimentaria, Instituto Universitario de Investigación de Carne y Productos Cárnicos, Facultad de Veterinaria, Universidad de Extremadura, 10003 Cáceres, Spain
| | - Josué Delgado
- Higiene y Seguridad Alimentaria, Instituto Universitario de Investigación de Carne y Productos Cárnicos, Facultad de Veterinaria, Universidad de Extremadura, 10003 Cáceres, Spain..
| | - María J Andrade
- Higiene y Seguridad Alimentaria, Instituto Universitario de Investigación de Carne y Productos Cárnicos, Facultad de Veterinaria, Universidad de Extremadura, 10003 Cáceres, Spain
| |
Collapse
|
16
|
Zheng D, Zou L, Zou J, Li Q, Lu S. Multi-omics analysis reveals potential mechanisms of diarrhetic shellfish toxin and fatty acid synthesis in marine harmful Prorocentrum. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137674. [PMID: 40007370 DOI: 10.1016/j.jhazmat.2025.137674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/31/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025]
Abstract
This study integrates transcriptomic and proteomic approaches to investigate the synthesis pathways of diarrhetic shellfish toxins (DSTs) in Prorocentrum lima and Prorocentrum arenarium, three strains exhibiting distinct toxin profiles. By combining multi-omics data, we identified 45 type I polyketide synthases (PKSs) and 45 type II fatty acid synthases (FASs) as potential candidates involved in DST production. Sequence analysis of the selected PKS and FAS genes revealed a high level of consistency across different omics datasets. Our results highlight the differential expression of proteins associated with fatty acid biosynthesis, with P. arenarium (HN231) exhibiting a significantly higher proportion of saturated fatty acids (SFAs) compared to P. lima (3XS36 and XS336), consistent with the upregulation of proteins involved in fatty acid synthesis pathways. These findings offer new insights into the molecular mechanisms underlying DST production and fatty acid metabolism in dinoflagellates, providing a foundation for future research on environmental contamination by DSTs. This study underscores the importance of multi-omics approaches for understanding hazardous marine toxins and their environmental implications.
Collapse
Affiliation(s)
- Danlin Zheng
- College of Life Science and Technology, and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Jinan University, Guangzhou 510362, China
| | - Ligong Zou
- College of Life Science and Technology, and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Jinan University, Guangzhou 510362, China
| | - Jian Zou
- College of Life Science and Technology, and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Jinan University, Guangzhou 510362, China
| | - Qun Li
- College of Life Science and Technology, and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Jinan University, Guangzhou 510362, China
| | - Songhui Lu
- College of Life Science and Technology, and Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Jinan University, Guangzhou 510362, China.
| |
Collapse
|
17
|
Gemeinhardt TM, Regy RM, Phan TM, Pal N, Sharma J, Senkovich O, Mendiola AJ, Ledterman HJ, Henrickson A, Lopes D, Kapoor U, Bihani A, Sihou D, Kim YC, Jeruzalmi D, Demeler B, Kim CA, Mittal J, Francis NJ. A disordered linker in the Polycomb protein Polyhomeotic tunes phase separation and oligomerization. Mol Cell 2025; 85:2128-2146.e15. [PMID: 40441156 PMCID: PMC12145237 DOI: 10.1016/j.molcel.2025.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 02/24/2025] [Accepted: 05/05/2025] [Indexed: 06/11/2025]
Abstract
Biomolecular condensates are increasingly recognized as key regulators of chromatin organization, yet how their formation and properties arise from protein sequences remains incompletely understood. Cross-species comparisons can reveal both conserved functions and significant evolutionary differences. Here, we integrate in vitro reconstitution, molecular dynamics simulations, and cell-based assays to examine how Drosophila and human variants of Polyhomeotic (Ph)-a subunit of the PRC1 chromatin regulatory complex-drive condensate formation through their sterile alpha motif (SAM) oligomerization domains. We identify divergent interactions between SAM and the disordered linker connecting it to the rest of Ph. These interactions enhance oligomerization and modulate both the formation and properties of reconstituted condensates. Oligomerization influences condensate dynamics but minimally impacts condensate formation. Linker-SAM interactions also affect condensate formation in Drosophila and human cells and growth in Drosophila imaginal discs. Our findings show how evolutionary changes in disordered linkers can fine-tune condensate properties, providing insights into sequence-function relationships.
Collapse
Affiliation(s)
- Tim M Gemeinhardt
- Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Roshan M Regy
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Tien M Phan
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Nanu Pal
- Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, Canada
| | - Jyoti Sharma
- Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada
| | - Olga Senkovich
- Department of Biochemistry and Molecular Genetics, Midwestern University, Glendale, AZ, USA
| | - Andrea J Mendiola
- Department of Biochemistry and Molecular Genetics, Midwestern University, Glendale, AZ, USA
| | - Heather J Ledterman
- Department of Biochemistry and Molecular Genetics, Midwestern University, Glendale, AZ, USA
| | - Amy Henrickson
- Department of Chemistry and Biochemistry, The University of Lethbridge, Lethbridge, AB, Canada
| | - Daniel Lopes
- Department of Chemistry and Biochemistry, City College of New York, New York, NY, USA
| | - Utkarsh Kapoor
- Department of Chemical and Biomedical Engineering, University of Wyoming, Laramie, WY, USA
| | - Ashish Bihani
- Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Djamouna Sihou
- Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, Canada
| | - Young C Kim
- Center for Materials Physics and Technology, Naval Research Laboratory, Washington, DC, USA
| | - David Jeruzalmi
- Department of Chemistry and Biochemistry, City College of New York, New York, NY, USA; Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, NY, USA; Ph.D. Program in Biology, The Graduate Center of the City University of New York, New York, NY, USA; Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, NY, USA
| | - Borries Demeler
- Department of Chemistry and Biochemistry, The University of Lethbridge, Lethbridge, AB, Canada; Department of Chemistry and Biochemistry, University of Montana, Missoula, MT, USA
| | - Chongwoo A Kim
- Department of Biochemistry and Molecular Genetics, Midwestern University, Glendale, AZ, USA
| | - Jeetain Mittal
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA; Department of Chemistry, Texas A&M University, College Station, TX, USA; Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
| | - Nicole J Francis
- Montreal Clinical Research Institute (IRCM), Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, Canada.
| |
Collapse
|
18
|
Srinivasan S, Ramos-Lewis W, Morais MR, Chi Q, Soh AW, Williams E, Lennon R, Sherwood DR. A collagen IV fluorophore knock-in toolkit reveals trimer diversity in C. elegans basement membranes. J Cell Biol 2025; 224:e202412118. [PMID: 40100062 PMCID: PMC11917169 DOI: 10.1083/jcb.202412118] [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: 12/14/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
The type IV collagen triple helix, composed of three ⍺-chains, is a core basement membrane (BM) component that assembles into a network within BMs. Endogenous tagging of all ⍺-chains with genetically encoded fluorophores has remained elusive, limiting our understanding of this crucial BM component. Through genome editing, we show that the C termini of the C. elegans type IV collagen ⍺-chains EMB-9 and LET-2 can be fused to a variety of fluorophores to create a strain toolkit with wild-type health. Using quantitative imaging, our results suggest a preference for LET-2-LET-2-EMB-9 trimer construction, but also tissue-specific flexibility in trimers assembled driven by differences in ⍺-chain expression levels. By tagging emb-9 and let-2 mutants that model human Gould syndrome, a complex multitissue disorder, we further discover defects in extracellular accumulation and turnover that might help explain disease pathology. Together, our findings identify a permissive tagging site in C. elegans that will allow diverse studies on type IV collagen regulation and function in animals.
Collapse
Affiliation(s)
| | | | - Mychel R.P.T. Morais
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Qiuyi Chi
- Department of Biology, Duke University, Durham, NC, USA
| | - Adam W.J. Soh
- Department of Biology, Duke University, Durham, NC, USA
| | - Emily Williams
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Rachel Lennon
- Division of Cell-Matrix Biology and Regenerative Medicine, Wellcome Centre for Cell-Matrix Research, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | | |
Collapse
|
19
|
Swensen AC, Piehowski PD, Chen J, Chan XY, Kelly SS, Petyuk VA, Moore RJ, Nasif L, Butterworth EA, Atkinson MA, Kulkarni RN, Campbell-Thompson M, Mathews CE, Qian WJ. Increased inflammation as well as decreased endoplasmic reticulum stress and translation differentiate pancreatic islets from donors with pre-symptomatic stage 1 type 1 diabetes and non-diabetic donors. Diabetologia 2025:10.1007/s00125-025-06417-3. [PMID: 40457096 DOI: 10.1007/s00125-025-06417-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/12/2025] [Indexed: 06/11/2025]
Abstract
AIMS/HYPOTHESIS Progression to type 1 diabetes is associated with genetic factors, the presence of autoantibodies and a decline in beta cell insulin secretion in response to glucose. Very little is known regarding the molecular changes that occur in human insulin-secreting beta cells prior to the onset of type 1 diabetes. Herein, we applied an unbiased proteomics approach to identify changes in proteins and potential mechanisms of islet dysfunction in islet-autoantibody-positive organ donors with pre-symptomatic stage 1 type 1 diabetes (HbA1c ≤42 mmol/mol [6.0%]). We aimed to identify pathways in islets that are indicative of beta cell dysfunction. METHODS Multiple islet sections were collected through laser microdissection of frozen pancreatic tissues from organ donors positive for single or multiple islet autoantibodies (AAb+, n=5), and age (±2 years)- and sex-matched non-diabetic (ND) control donors ( n=5) obtained from the Network for Pancreatic Organ donors with Diabetes (nPOD). Islet sections were subjected to MS-based proteomics and analysed with label-free quantification followed by pathway and functional annotations. RESULTS Analyses resulted in ~4500 proteins identified with low false discovery rate (<1%), with 2165 proteins reliably quantified in every islet sample. We observed large inter-donor variations that presented a challenge for statistical analysis of proteome changes between donor groups. We therefore focused on only the donors with stage 1 type 1 diabetes who were positive for multiple autoantibodies (mAAb+, n=3) and genetic risk compared with their matched ND controls (n=3) for the final statistical analysis. Approximately 10% of the proteins (n=202) were significantly different (unadjusted p<0.025, q<0.15) for mAAb+ vs ND donor islets. The significant alterations clustered around major functions for upregulation in the immune response and glycolysis, and downregulation in endoplasmic reticulum (ER) stress response as well as protein translation and synthesis. The observed proteome changes were further supported by several independent published datasets, including a proteomics dataset from in vitro proinflammatory cytokine-treated human islets and single-cell RNA-seq datasets from AAb+ individuals. CONCLUSIONS/INTERPRETATION In situ human islet proteome alterations in stage 1 type 1 diabetes centred around several major functional categories, including an expected increase in immune response genes (elevated antigen presentation/HLA), with decreases in protein synthesis and ER stress response, as well as compensatory metabolic response. The dataset serves as a proteomics resource for future studies on beta cell changes during type 1 diabetes progression and pathogenesis. DATA AVAILABILITY The LC-MS raw datasets that support the findings of this study have been deposited in the online repository: MassIVE ( https://massive.ucsd.edu/ProteoSAFe/static/massive.jsp ) with accession no. MSV000090212.
Collapse
Affiliation(s)
- Adam C Swensen
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Paul D Piehowski
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Jing Chen
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
- Department of Infectious Disease and Immunology, University of Florida, Gainesville, FL, USA
| | - X'avia Y Chan
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Shane S Kelly
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Vladislav A Petyuk
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Ronald J Moore
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Lith Nasif
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Elizabeth A Butterworth
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Rohit N Kulkarni
- Section of Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center; Department of Medicine, Beth Israel Deaconess Medical Center; Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Clayton E Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, USA.
- Department of Infectious Disease and Immunology, University of Florida, Gainesville, FL, USA.
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, WA, USA.
| |
Collapse
|
20
|
Jaffray EG, Tatham MH, Mojsa B, Plechanovová A, Rojas-Fernandez A, Liu JC, Mailand N, Ibrahim AF, Ball G, Porter IM, Hay RT. PML mutants from arsenic-resistant patients reveal SUMO1-TOPORS and SUMO2/3-RNF4 degradation pathways. J Cell Biol 2025; 224:e202407133. [PMID: 40239066 PMCID: PMC12002637 DOI: 10.1083/jcb.202407133] [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: 07/20/2024] [Revised: 01/31/2025] [Accepted: 03/10/2025] [Indexed: 04/18/2025] Open
Abstract
Arsenic effectively treats acute promyelocytic leukemia by inducing SUMO and ubiquitin-dependent degradation of the promyelocytic leukemia (PML)-retinoic acid receptor alpha oncogenic fusion protein. However, some patients relapse with arsenic-resistant disease because of missense mutations in PML. To determine the mechanistic basis for arsenic resistance, PML-/- cells were reconstituted with YFP fusions of wild-type PML-V and two common patient mutants: A216T and L217F. Both mutants were resistant to degradation by arsenic but for different biochemical reasons. Arsenic did not trigger SUMOylation of A216T PML, which failed to recruit the SUMO-targeting ubiquitin ligases RNF4 and TOPORS. L217F PML did respond with increased SUMO2/3 conjugation that facilitated RNF4 engagement but failed to reach the threshold of SUMO1 conjugation required to recruit TOPORS. Thus, neither mutant accumulated the appropriate polyubiquitin signal required for p97 binding. These PML mutants have revealed a convergence of SUMO1, SUMO2/3, TOPORS, and RNF4 that facilitates the arsenic-induced degradation of PML.
Collapse
Affiliation(s)
- Ellis G. Jaffray
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Michael H. Tatham
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Barbara Mojsa
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Anna Plechanovová
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | | | - Julio C.Y. Liu
- Protein Signaling Program, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Niels Mailand
- Protein Signaling Program, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Adel F.M. Ibrahim
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| | - Graeme Ball
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, Dundee, UK
| | | | - Ronald T. Hay
- Division of Molecular, Cell and Developmental Biology, School of Life Sciences, University of Dundee, Dundee, UK
| |
Collapse
|
21
|
Ouchi K, Masuda T, Yonemaru K, Isono K, Ohya Y, Shiraki N, Tasaki M, Inomata Y, Ueda M, Era T, Kume S, Ando Y, Jono H. Dynamic changes of intracellular signals in ATTR Tyr114Cys amyloidosis. Biochem Biophys Rep 2025; 42:102012. [PMID: 40275963 PMCID: PMC12020847 DOI: 10.1016/j.bbrep.2025.102012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
Hereditary transthyretin (TTR) amyloidosis (ATTRv amyloidosis) is an autosomal dominant disease caused by various TTR mutations. Despite the fact that ATTR Tyr114Cys (p.Tyr134Cys) amyloidosis (tyrosine to cysteine at codon 114) exhibits poorer prognosis than other ATTRv amyloidosis and leads to death due to severe clinical symptoms, the molecular pathogenesis of ATTR Tyr114Cys amyloidosis is still largely unknown. In this study, we took advantage of ATTR Tyr114Cys amyloidosis-specific induced pluripotent stem (iPS) cells to differentiate into hepatocyte-like cells (Y114C-HLCs), which are mainly TTR producing cells, and elucidated their pathogenesis. We performed proteomic analysis to comprehensively identify specific intracellular signaling pathways involved in Y114C-HLCs, and identified the specific proteins changed only in Y114C-HLCs, in comparison with disease control HLCs from ATTR Val30Met amyloidosis (V30M-HLCs). Moreover, we have succeeded in identifying several specific intracellular signals that are significantly activated in Y114C-HLCs, including cellular responses to stress and extracellular matrix organization. Our proteomic analysis is the first to report that the specific point mutations in ATTRv amyloidosis cause dynamic changes in cellular response, and reveal the specific intracellular signals may be involved in the specific pathogenesis of ATTR Tyr114Cys amyloidosis.
Collapse
Affiliation(s)
- Kenta Ouchi
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| | - Takeshi Masuda
- Graduate School of Media and Governance / Institute for Advanced Biosciences, Keio University, 14-1 Baba-machi, Tsuruoka-City, Yamagata Prefecture, 997-0035, Japan
| | - Kou Yonemaru
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| | - Kaori Isono
- Department of Transplantation and Paediatric Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| | - Yuki Ohya
- Department of Transplantation and Paediatric Surgery, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
- Department of Pediatric Surgery, Kumamoto Rosai Hospital, 1670 Takehara-cho, Yatsushiro City, Kumamoto Prefecture, 866-0826, Japan
| | - Nobuaki Shiraki
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori Ward, Yokohama City, Kanagawa Prefecture, 226-8501, Japan
| | - Masayoshi Tasaki
- Department of Biomedical Laboratory Sciences, Graduate School of Health Sciences, Kumamoto University, Kumamoto, 1-1-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
- Department of Neurology, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| | - Yukihiro Inomata
- Department of Pediatric Surgery, Kumamoto Rosai Hospital, 1670 Takehara-cho, Yatsushiro City, Kumamoto Prefecture, 866-0826, Japan
| | - Mitsuharu Ueda
- Department of Neurology, Graduate School of Medical Science, Kumamoto University, 1-1-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| | - Shoen Kume
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori Ward, Yokohama City, Kanagawa Prefecture, 226-8501, Japan
| | - Yukio Ando
- Department of Amyloidosis Research, Nagasaki International University, 2825-7 Huis Ten Bosch Cho, Sasebo City, Nagasaki Prefecture, 859-3298, Japan
| | - Hirofumi Jono
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
- Department of Pharmacy, Kumamoto University Hospital, 1-1-1 Honjo, Chuo Ward, Kumamoto City, Kumamoto Prefecture, 860-8556, Japan
| |
Collapse
|
22
|
Pang Y, Wu L, Xia J, Xu X, Gao C, Hou L, Jiang L. Trim38 attenuates pressure overload‑induced cardiac hypertrophy by suppressing the TAK1/JNK/P38 signaling pathway. Int J Mol Med 2025; 55:98. [PMID: 40314083 PMCID: PMC12045468 DOI: 10.3892/ijmm.2025.5539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 02/20/2025] [Indexed: 05/03/2025] Open
Abstract
Pathological cardiac hypertrophy is a major contributor to heart failure (HF), resulting in high mortality rates worldwide; therefore, identifying key molecules in pathological cardiac hypertrophy is of critical importance for preventing or reversing HF. Tripartite motif 38 (Trim38) is an E3 ubiquitin ligase that serves a pivotal role in various diseases. The present study aimed to elucidate the regulatory role of Trim38 in pressure overload‑induced pathological cardiac hypertrophy and to explore its underlying molecular mechanisms. The expression of Trim38 was decreased in hypertrophic heart tissues from a murine model of transverse aortic constriction (TAC) and in neonatal rat cardiomyocytes (NRCMs) treated with phenylephrine (PE). Furthermore, Trim38 knockout (Trim38‑KO) aggravated cardiac hypertrophy after TAC, and Trim38 knockdown in cardiomyocytes increased cell cross section area, and upregulated the expression of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP) following treatment with PE. Ubiquitinomics analysis revealed that the MAPK signaling pathway was regulated by Trim38. Furthermore, western blotting confirmed that Trim38‑KO activated TAK1 and JNK/P38. By contrast, Trim38 overexpression in NRCMs suppressed the JNK/P38 signaling pathway and inhibited the phosphorylation of TAK1. Furthermore, Trim38 knockdown resulted in a marked enhancement of TAK1 phosphorylation, concomitant with an augmentation of cardiomyocyte area and a significant upregulation of the hypertrophic biomarkers ANP and BNP. By contrast, infection with an adenovirus containing dominant‑negative TAK1 inhibited TAK1 activity, which attenuated Trim38 knockdown‑induced cardiomyocyte hypertrophy, confirming that TAK1 is a key molecule involved in the protective effects of Trim38 on cardiomyocytes. In conclusion, to the best of our knowledge, the present study is the first to reveal that Trim38 confers protection against pathological cardiac hypertrophy by inhibiting the TAK1/JNK/P38 signaling pathway; therefore, Trim38 may be a promising target for treating cardiac hypertrophy.
Collapse
Affiliation(s)
- Yanan Pang
- Institute of Cardiovascular Diseases, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
- Department of Cardiology, Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 201600, P.R. China
| | - Luyao Wu
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Jiachun Xia
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Xin Xu
- Collaborative Innovation Centre of Regenerative Medicine and Medical Bioresource Development and Application Co-constructed by The Province and Ministry, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Chenshan Gao
- Division of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| | - Lei Hou
- Department of Cardiology, Songjiang Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 201600, P.R. China
| | - Li Jiang
- Institute of Cardiovascular Diseases, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, P.R. China
| |
Collapse
|
23
|
Jabbar J, Afroze B, Ling NXY, Oakhill JS, Rouiller I. Lysine acetylation modulates s-OPA1 GTPase activity and oligomerization in mitochondrial membrane remodeling. Protein Sci 2025; 34:e70179. [PMID: 40437978 PMCID: PMC12120360 DOI: 10.1002/pro.70179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 06/01/2025]
Abstract
Mitochondrial dynamics are regulated by coordinated fission and fusion events that rely on key proteins and lipids organized spatially within the mitochondria. The dynamin-related GTPase Optic Atrophy 1 (OPA1) is essential for inner mitochondrial membrane fusion and cristae structure maintenance. While post-translational modifications, particularly lysine acetylation, are emerging as critical regulators of mitochondrial protein function, their impact on OPA1 remains poorly characterized. In this study, we explored the effects of lysine acetylation on the short form of OPA1 (s-OPA1) using acetylation and deacetylation mimetic mutations. Through a combination of in silico analyses and functional assays, we identified lysine residues in s-OPA1 that are conserved across species and significantly influence protein stability, GTPase activity, and oligomeric assembly upon acetylation or deacetylation. Our findings reveal that acetylation at K328 and deacetylation at K342 within the G domain enhance the GTPase activity of s-OPA1 upon lipid membrane binding, whereas deacetylation at K772 abolishes membrane binding-induced GTPase activity. Negative-stain transmission electron microscopy indicated that while lysine acetylation does not alter the ability of s-OPA1 to bind and tubulate liposomes, it significantly impacts higher-order filament formation. These findings provide novel insights into how acetylation modulates s-OPA1 function, highlighting a potential mechanism for post-translational regulation of mitochondrial dynamics. Our study contributes to the understanding of how molecular changes influence broader cellular processes, with implications for mitochondrial function and related disorders.
Collapse
Affiliation(s)
- Javaid Jabbar
- Department of Biochemistry & PharmacologyBio21 Molecular Science and Biotechnology Institute, University of MelbourneParkvilleVictoriaAustralia
- ARC Centre for Cryo‐electron Microscopy of Membrane ProteinsParkvilleVictoriaAustralia
| | - Bakht Afroze
- Department of Biochemistry & PharmacologyBio21 Molecular Science and Biotechnology Institute, University of MelbourneParkvilleVictoriaAustralia
| | - Naomi X. Y. Ling
- St. Vincent's Institute of Medical ResearchFitzroyVictoriaAustralia
| | - Jonathan S. Oakhill
- St. Vincent's Institute of Medical ResearchFitzroyVictoriaAustralia
- Department of MedicineUniversity of MelbourneParkvilleVictoriaAustralia
- Faculty of Health SciencesAustralian Catholic UniversityMelbourneVictoriaAustralia
| | - Isabelle Rouiller
- Department of Biochemistry & PharmacologyBio21 Molecular Science and Biotechnology Institute, University of MelbourneParkvilleVictoriaAustralia
- ARC Centre for Cryo‐electron Microscopy of Membrane ProteinsParkvilleVictoriaAustralia
| |
Collapse
|
24
|
Valentim-Coelho C, Saraiva J, Osório H, Antunes M, Vaz F, Neves S, Pinto P, Bárbara C, Penque D. Red blood cell proteomic profiling in mild and severe obstructive sleep apnea patients before and after positive airway pressure treatment. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167767. [PMID: 40043591 DOI: 10.1016/j.bbadis.2025.167767] [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: 07/01/2024] [Revised: 01/05/2025] [Accepted: 02/25/2025] [Indexed: 04/15/2025]
Abstract
Obstructive Sleep Apnea (OSA) is characterized by recurrent-episodes of apneas/hypopneas during sleep, leading to recurrent intermittent-hypoxia and sleep fragmentation. Non-treated OSA can result in cardiometabolic diseases. In this study, we applied a shotgun-proteomics strategy to deeper investigate the red blood cell-(RBC) homeostasis regulation in the context of OSA-severity and their response to six months of positive airway pressure (PAP)-treatment. RBC-samples from patients with Mild/Severe-OSA before/after-PAP treatment and patients as simple-snoring controls were selected. The mass-spectrometry raw-data was analysed by MaxQuant for protein identification/quantification followed by statistical Linear Models-(LM) and Linear Mixed Models-(LMM) to investigate OSA-severity effect and interaction with PAP, respectively. The functional/biological network analysis were performed by DAVID-platform. The results indicated that key-enzymes of the Embden-Meyerhof-Parnas (EMP)-glycolysis and pentose phosphate pathway-(PPP) were differentially changed in Severe-OSA, suggesting that the O2-dependent metabolic flux through EMP and PPP maybe compromised in these cells due to severe intermittent hypoxia/reoxygenation-induced oxidative-stress events in these patients. The Rapoport-Luebering-glycolytic shunt showed a significant downregulation across OSA-severity maybe to increase hemoglobin-O2 affinity to adapt to O2 low availability in the lung, although EMP-glycolysis showed decreased only in Severe-OSA. Proteins of the immunoproteasome were upregulated in Severe-OSA maybe to respond to severe oxidative-stress. In Mild-OSA, proteins related to the ubiquitination/neddylation-(Ub/Ned)-dependent proteasome system were upregulated. After PAP, proteins of Glycolysis and Ub/Ned-dependent proteasome system showed reactivated in Severe-OSA. In Mild-OSA, PAP induced upregulation of immunoproteasome proteins, suggesting that this treatment may increase oxidative-stress in these patients. Once validated these proteins maybe candidate biomarkers for OSA or OSA-therapy response.
Collapse
Affiliation(s)
- Cristina Valentim-Coelho
- Laboratório de Proteómica, Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge - INSA, 1649-016 Lisboa, Portugal; Centro de Toxicogenómica e Saúde Humana (ToxOmics), Comprehensive Health Research Center (CHRC), Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal.
| | - Joana Saraiva
- Laboratório de Proteómica, Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge - INSA, 1649-016 Lisboa, Portugal; Centro de Toxicogenómica e Saúde Humana (ToxOmics), Comprehensive Health Research Center (CHRC), Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Hugo Osório
- Instituto de Investigação e Inovação em Saúde - i3S, Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Patologia e Imunologia Molecular da Universidade do Porto - Ipatimup, 4200-135 Porto, Portugal; Departamento de Patologia, Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
| | - Marília Antunes
- Centro de Estatística e Aplicações da Universidade de Lisboa e Departamento de Estatística e Investigação Operacional, Faculdade de Ciências, Universidade de Lisboa, 1749-016 Lisboa, Portugal
| | - Fátima Vaz
- Laboratório de Proteómica, Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge - INSA, 1649-016 Lisboa, Portugal; Centro de Toxicogenómica e Saúde Humana (ToxOmics), Comprehensive Health Research Center (CHRC), Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Sofia Neves
- Laboratório de Proteómica, Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge - INSA, 1649-016 Lisboa, Portugal; Centro de Toxicogenómica e Saúde Humana (ToxOmics), Comprehensive Health Research Center (CHRC), Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal
| | - Paula Pinto
- Serviço de Pneumologia, Centro Hospitalar Lisboa Norte - CHLN, 1649-035 Lisboa, Portugal; Instituto de Saúde Ambiental - ISAMB, Faculdade de Medicina, Universidade de Lisboa, 1649-026 Lisboa, Portugal
| | - Cristina Bárbara
- Serviço de Pneumologia, Centro Hospitalar Lisboa Norte - CHLN, 1649-035 Lisboa, Portugal; Instituto de Saúde Ambiental - ISAMB, Faculdade de Medicina, Universidade de Lisboa, 1649-026 Lisboa, Portugal
| | - Deborah Penque
- Laboratório de Proteómica, Departamento de Genética Humana, Instituto Nacional de Saúde Dr. Ricardo Jorge - INSA, 1649-016 Lisboa, Portugal; Centro de Toxicogenómica e Saúde Humana (ToxOmics), Comprehensive Health Research Center (CHRC), Universidade Nova de Lisboa, 1150-082 Lisboa, Portugal.
| |
Collapse
|
25
|
Monticelli M, Paris D, Monti MC, Morretta E, Pakanova Z, Nemcovic M, Kodrikova R, Cubellis MV, Andreotti G. Beneficial effects of Glc-1,6-P 2 modulation on mutant phosphomannomutase-2. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119948. [PMID: 40169095 DOI: 10.1016/j.bbamcr.2025.119948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/18/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
The metabolite Glucose-1,6-bisphosphate (Glc-1,6-P2) plays a vital role in human metabolism, and is a crucial activator and stabilizer for phosphomannomutase-2 (PMM2) - mutations within this protein propagate the most common congenital disorder of glycosylation (PMM2-CDG). In vivo, Glc-1,6-P2 is hydrolysed by phosphomannomutase-1 (PMM1), predominantly in the brain, under the influence of inosine monophosphate (IMP). In the present study, we employed knock-out PMM1 in Arg141His/Phe119LeuPMM2 patient-derived fibroblasts and investigated the phenotypic improvement. Increased Glc-1,6-P2 was associated with glycosylation enhancement, confirmed by glycan profiling. Previously identified PMM2-CDG biomarkers, such as LAMP1, PTX3 and lysosomal enzymes showed empirical imrovement- these findings were corroborated by metabolomic and proteomic analysis. Moreover, our results support the potential of Glc-1,6-P2 modulation for PMM2-CDG, potentiating novel perspectives in drug discovery.
Collapse
Affiliation(s)
- Maria Monticelli
- Institute of Biomolecular Chemistry, National Research Council of Italy, Comprensorio Olivetti, via Campi Flegrei 34, 80078 Pozzuoli, Italy; Dept. Biology, University of Napoli "Federico II", Complesso Universitario Monte Sant'Angelo, via Cinthia, 80126 Naples, Italy
| | - Debora Paris
- Institute of Biomolecular Chemistry, National Research Council of Italy, Comprensorio Olivetti, via Campi Flegrei 34, 80078 Pozzuoli, Italy
| | - Maria Chiara Monti
- Department of Pharmacy, University of Napoli "Federico II", via Tommaso De Amicis 95, 80131 Naples, Italy
| | - Elva Morretta
- Department of Pharmacy, University of Napoli "Federico II", via Tommaso De Amicis 95, 80131 Naples, Italy
| | - Zuzana Pakanova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 84538 Bratislava, Slovakia
| | - Marek Nemcovic
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 84538 Bratislava, Slovakia
| | - Rebeka Kodrikova
- Institute of Chemistry, Slovak Academy of Sciences, Dubravska cesta 9, 84538 Bratislava, Slovakia
| | - Maria Vittoria Cubellis
- Dept. Biology, University of Napoli "Federico II", Complesso Universitario Monte Sant'Angelo, via Cinthia, 80126 Naples, Italy
| | - Giuseppina Andreotti
- Institute of Biomolecular Chemistry, National Research Council of Italy, Comprensorio Olivetti, via Campi Flegrei 34, 80078 Pozzuoli, Italy.
| |
Collapse
|
26
|
Hung WT, Tsou KC, Cho HC, Wu PS, Lin MH, Chen SC, Liao HC, Lu CW, Li CF, Su WC, Huang CH, Hsu WM, Ju YT, Tu CF, Lin SJ, Hsu HH, Chen JS, Young TH. Chondrogenic Potential of Cryopreserved Aortic Allografts: Guiding Perichondrial Regeneration in Tracheal Repair. Adv Healthc Mater 2025; 14:e2405106. [PMID: 40357702 DOI: 10.1002/adhm.202405106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/08/2025] [Indexed: 05/15/2025]
Abstract
Native tracheal cartilage exhibits limited regenerative capacity, making the search for suitable biomaterials for tracheal repair a persistent challenge. In this study, a non-decellularized cryopreserved aortic allograft (CAo) is investigated as a scaffold for tracheal cartilage regeneration. Originally used to reconstruct infected aortas, CAo retains key features of a large artery-abundant elastic fibers and smooth muscle cells-and demonstrates favorable in vitro biocompatibility with chondrocytes. A trachea-CAo patch construct maintains tensile properties comparable to native trachea and tolerates normal expiratory forces. In a rabbit patch-defect model, CAo elicits only a mild-to-moderate immune response that gradually subsides. Within one month of implantation, robust neocartilage formation is observed, along with angiogenesis and epithelial regeneration. Over the next 12 months, the original aortic scaffold progressively degrades, while newly formed cartilage-originating from recipient perichondrial chondroprogenitor cells-replaces it. Proteomic analyses show that CAo is enriched in cytoskeletal, adhesion, cell migration, and extracellular matrix (ECM)-related proteins, with fibroblast growth factor 2 emerging as a critical mediator of chemotaxis and chondrogenic differentiation. These findings indicate that CAo serves as both a structural and biological scaffold, activating tracheal cartilage regeneration through synergistic biocompatibility, growth factor signaling, and ECM support.
Collapse
Affiliation(s)
- Wan-Ting Hung
- Department of Surgery, National Taiwan University Hospital, No. 7, Zhongshan S. Rd, Taipei, 100225, Taiwan (R.O.C.)
- Department of Surgery, College of Medicine, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Rm733, Bldg.Lab.Med., NTU Hospital, No.1, Chang-Te St., Taipei, 100229, Taiwan (R.O.C.)
| | - Kuan-Chuan Tsou
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Rm733, Bldg.Lab.Med., NTU Hospital, No.1, Chang-Te St., Taipei, 100229, Taiwan (R.O.C.)
- Division of Thoracic Surgery, Department of Surgery, Taipei City Hospital Zhongxiao Branch, No.87, Tongde Rd., Nangang Dist., Taipei, 115006, Taiwan (R.O.C.)
| | - Huan-Chieh Cho
- NTU Consortium of Integrative Biomedical Science Key Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 106319, Taiwan (R.O.C.)
| | - Pei-Shan Wu
- Department of Microbiology, College of Medicine, National Taiwan University, No.1, Sec., 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
| | - Miao-Hsia Lin
- Department of Microbiology, College of Medicine, National Taiwan University, No.1, Sec., 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
| | - Sy-Chi Chen
- Department of Surgery, National Taiwan University Hospital, No. 7, Zhongshan S. Rd, Taipei, 100225, Taiwan (R.O.C.)
| | - Hsien-Chi Liao
- Department of Surgery, College of Medicine, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Rm733, Bldg.Lab.Med., NTU Hospital, No.1, Chang-Te St., Taipei, 100229, Taiwan (R.O.C.)
- Department of Traumatology, National Taiwan University Hospital, No. 7, Zhongshan S. Rd, Taipei, 100225, Taiwan (R.O.C.)
| | - Chao-Wen Lu
- Department of Surgery, National Taiwan University Hospital, No. 7, Zhongshan S. Rd, Taipei, 100225, Taiwan (R.O.C.)
| | - Chi-Fang Li
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
| | - Wei-Ching Su
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
| | - Chih-Hsuan Huang
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, No. 7, Zhongshan S. Rd, Taipei, 100225, Taiwan (R.O.C.)
- Department of Surgery, College of Medicine, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
| | - Yu-Ten Ju
- Department of Animal Science and Technology, National Taiwan University, No. 50, Ln. 155, Sec. 3, Keelung Rd., Taipei, 106326, Taiwan (R.O.C.)
| | - Ching-Fu Tu
- Division of Animal Technology, Animal Technology Laboratories, Agricultural Technology Research Institute, No.1, Ln. 51, Dahu Rd., Xiangshan Dist., Hsinchu City, 300110, Taiwan (R.O.C.)
| | - Sung-Jan Lin
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, No.1, Sec., 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Rm. 625, No. 49, Fanglan Rd., Da'an Dist., Taipei City, 106038, Taiwan (R.O.C.)
| | - Hsao-Hsun Hsu
- Department of Surgery, National Taiwan University Hospital, No. 7, Zhongshan S. Rd, Taipei, 100225, Taiwan (R.O.C.)
- Department of Surgery, College of Medicine, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
- Department of Surgical Oncology, National Taiwan University Cancer Center, No.57, Ln. 155, Sec. 3, Keelung Rd., Taipei, 106326, Taiwan (R.O.C.)
| | - Jin-Shing Chen
- Department of Surgery, National Taiwan University Hospital, No. 7, Zhongshan S. Rd, Taipei, 100225, Taiwan (R.O.C.)
- Department of Surgery, College of Medicine, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
- Department of Surgical Oncology, National Taiwan University Cancer Center, No.57, Ln. 155, Sec. 3, Keelung Rd., Taipei, 106326, Taiwan (R.O.C.)
| | - Tai-Horng Young
- Department of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No.1, Sec. 1, Jen Ai Rd., Taipei, 100233, Taiwan (R.O.C.)
| |
Collapse
|
27
|
Sharma N, Sharma N, Biswas A, Gupta S, Behura A, Rodriguez GM. Iron-restricted Mycobacterium tuberculosis exports pathogenicity factors packed in extracellular vesicles. PLoS One 2025; 20:e0324919. [PMID: 40445943 PMCID: PMC12124568 DOI: 10.1371/journal.pone.0324919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
Mycobacterium tuberculosis, the pathogen responsible for human tuberculosis, responds to iron limitation by increasing the production of extracellular vesicles. This study examined the protein composition of induced M. tuberculosis extracellular membrane vesicles using chromatography coupled with mass spectrometry. The results revealed that vesicles contain key pathogenicity factors, including proteins that enhance bacterial survival, immune evasion, and inflammation. These findings deepen our understanding of the potential role of extracellular vesicles in M. tuberculosis-host interactions. The data can also aid in identifying new biomarkers of infection and developing vesicle-based, culture-independent TB diagnostic platforms.
Collapse
Affiliation(s)
- Nishant Sharma
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Nevadita Sharma
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Ashis Biswas
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Shamba Gupta
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Assirbad Behura
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Gloria Marcela Rodriguez
- The Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| |
Collapse
|
28
|
Sin YC, Abernathy B, Yuan ZF, Heier JL, Gonzalez JE, Parker LL, Mashek DG, Chen Y. Sorbate induces lysine sorbylation through noncanonical activities of class I HDACs to regulate the expression of inflammation genes. SCIENCE ADVANCES 2025; 11:eadv1071. [PMID: 40446041 PMCID: PMC12124360 DOI: 10.1126/sciadv.adv1071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 04/25/2025] [Indexed: 06/02/2025]
Abstract
Environmental factors may affect gene expression through epigenetic modifications of histones and transcription factors. Here, we report that cellular uptake of sorbate, a common food preservative, induces lysine sorbylation (Ksor) in mammalian cells and tissue mediated by the noncanonical activities of class I histone deacetylases (HDAC1-3). We demonstrated that HDAC1-3 catalyze sorbylation upon sorbate uptake and desorbylation in the absence of sorbate both in vitro and in cells. Sorbate uptake in mice livers significantly induced histone Ksor, correlating with decreased expressions of inflammation-response genes. Accordingly, sorbate treatment in macrophage RAW264.7 cells upon lipopolysaccharide (LPS) stimulation dose-dependently down-regulated proinflammatory gene expressions and nitric oxide production. Proteomic profiling identified RelA, a component of the NF-κB complex, and its interacting proteins as bona fide Ksor targets and sorbate treatment significantly decreased NF-κB transcriptional activities in response to LPS stimulation in RAW264.7 cells. Together, our study demonstrated a noncanonical mechanism of sorbate uptake in regulating epigenetic histone modifications and inflammatory gene expression.
Collapse
Affiliation(s)
- Yi-Cheng Sin
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
- Bioinformatics and Computational Biology Program, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Breann Abernathy
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Zuo-fei Yuan
- Center for Proteomics and Metabolomics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Jason L. Heier
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Justin E. Gonzalez
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Laurie L. Parker
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Douglas G. Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, University of Minnesota Twin Cities, Minneapolis, MN, USA
- Institute for the Biology of Aging and Metabolism, University of Minnesota Twin Cities, Minneapolis, MN, USA
| | - Yue Chen
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota Twin Cities, Minneapolis, MN, USA
| |
Collapse
|
29
|
Bebek G, Miyagi M, Wang X, Appleby BS, Leverenz JB, Pillai JA. Protein co-aggregates of dense core amyloid plaques and CSF differ in rapidly progressive Alzheimer's disease and slower sporadic Alzheimer's disease. Alzheimers Res Ther 2025; 17:118. [PMID: 40420296 PMCID: PMC12107742 DOI: 10.1186/s13195-025-01767-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND The rapidly progressive phenotype of Alzheimer's disease (rpAD) remains a rare and less-studied entity. Therefore, the replication of key results from the rpAD brain and cerebrospinal fluid (CSF) is lacking. METHODS A label-free quantitative LC-MS/MS analysis of proteins co-aggregating with core-amyloid β plaques in fresh frozen tissue (FFT) from medial temporal regions of rpAD ( n = 8 ) neuropathologically characterized at the National Prion Disease Pathology Surveillance Center (NPDPSC), compared with microdissected amyloid plaques from formalin-fixed, paraffin-embedded (FFPE) tissue blocks from patients with rpAD ( n = 22 ) previously published from the NPDPSC cohort, was performed. Matched rpAD CSF from the FFT cases were compared to a previously published proteomic evaluation of CSF in the AD subtype with rapid progression. RESULTS A total of 1841 proteins were characterized in the FFT study, of which 463 were consistently identified in every rpAD patient analyzed. One thousand two hundred eighty-three proteins were shared between the FFT and the prior FFPE study. FFT offered a more comprehensive proteomic profile than the prior FFPE study and prominently included the immune system pathways. Thirty-five proteins were shared in the FFT brain tissue, matched CSF from the same subjects, in which biological processes related to immune response were again notable. These results were validated against prior published proteomic CSF AD data with a faster rate of progression to identify the top 5 potential protein biomarkers of rapid progression in AD CSF. CONCLUSIONS These results support a distinct immune-related proteomic profile in both the brain and the CSF that can be explored as potential biomarkers in the future for the clinical diagnosis of rpAD.
Collapse
Affiliation(s)
- Gurkan Bebek
- Center for Proteomics and Bioinformatics, Department of Nutrition, Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Masaru Miyagi
- Department of Pharmacology, Case Western Reserve University, Cleveland, 44106, OH, USA
| | - Xinglong Wang
- Department of Pathology, University of Arizona, Tucson, 85721, AZ, USA
| | - Brian S Appleby
- Department of Neurology, National Prion Disease Pathology Surveillance Center, University Hospitals Cleveland Medical Center, Cleveland, 44195, OH, USA
| | - James B Leverenz
- Lou Ruvo Center for Brain Health, Neurological Institute, Department of Neurology, Cleveland, OH, USA
| | - Jagan A Pillai
- Lou Ruvo Center for Brain Health, Neurological Institute, Department of Neurology, Cleveland, OH, USA.
| |
Collapse
|
30
|
Itang ECM, Albrecht V, Schebesta AS, Thielert M, Lanz AL, Danhauser K, Jin J, Prell T, Strobel S, Klein C, Mann M, Pangratz-Fuehrer S, Mueller-Reif J. Ontology-guided clustering enables proteomic analysis of rare pediatric disorders. EMBO Mol Med 2025:10.1038/s44321-025-00253-z. [PMID: 40425748 DOI: 10.1038/s44321-025-00253-z] [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: 12/18/2024] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
The study of rare pediatric disorders is fundamentally limited by small patient numbers, making it challenging to draw meaningful biological conclusions. To address this, we developed a framework integrating clinical ontologies with proteomic profiling, enabling the systematic analysis of rare conditions in aggregate. We applied this approach to urine and plasma samples from 1140 children and adolescents, encompassing 394 distinct disease conditions and healthy controls. Using advanced mass spectrometry workflows, we quantified over 5000 proteins in urine, 900 in undepleted (neat) plasma, and 1900 in perchloric acid-depleted plasma. Embedding SNOMED CT clinical terminology in a network structure allowed us to group rare conditions based on their clinical relationships, enabling statistical analysis even for diseases with as few as two patients. This approach revealed molecular signatures across developmental stages and disease clusters while accounting for age- and sex-specific variation. Our framework provides a generalizable solution for studying heterogeneous patient populations where traditional case-control studies are impractical, bridging the gap between clinical classification and molecular profiling of rare diseases.
Collapse
Affiliation(s)
- Ericka C M Itang
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site Munich, Munich, Germany
| | - Vincent Albrecht
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Alicia-Sophie Schebesta
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
- German Center for Child and Adolescent Health (DZKJ), partner site Munich, Munich, Germany
| | - Marvin Thielert
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Anna-Lisa Lanz
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Katharina Danhauser
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Jessica Jin
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Tobias Prell
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sophie Strobel
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Christoph Klein
- German Center for Child and Adolescent Health (DZKJ), partner site Munich, Munich, Germany
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Susanne Pangratz-Fuehrer
- German Center for Child and Adolescent Health (DZKJ), partner site Munich, Munich, Germany
- Department of Pediatrics, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Johannes Mueller-Reif
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany.
- German Center for Child and Adolescent Health (DZKJ), partner site Munich, Munich, Germany.
| |
Collapse
|
31
|
Tian SZ, Yang Y, Ning D, Yu T, Gao T, Deng Y, Fang K, Xu Y, Jing K, Huang G, Chen G, Yin P, Li Y, Zeng F, Tian R, Zheng M. Landscape of the Epstein-Barr virus-host chromatin interactome and gene regulation. EMBO J 2025:10.1038/s44318-025-00466-5. [PMID: 40425856 DOI: 10.1038/s44318-025-00466-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
The three-dimensional (3D) chromatin structure of Epstein-Barr virus (EBV) within host cells and the underlying mechanisms of chromatin interaction and gene regulation, particularly those involving EBV's noncoding RNAs (ncRNAs), have remained incompletely characterized. In this study, we employed state-of-the-art techniques of 3D genome mapping, including protein-associated chromatin interaction analysis with paired-end tag sequencing (ChIA-PET), RNA-associated chromatin interaction technique (RDD), and super-resolution microscopy, to delineate the spatial architecture of EBV in human lymphoblastoid cells. We systematically analyzed EBV-to-EBV (E-E), EBV-to-host (E-H), and host-to-host (H-H) interactions linked to host proteins and EBV RNAs. Our findings reveal that EBV utilizes host CCCTC-binding factor (CTCF) and RNA polymerase II (RNAPII) to form distinct chromatin contact domains (CCDs) and RNAPII-associated interaction domains (RAIDs). The anchors of these chromatin domains serve as platforms for extensive interactions with host chromatin, thus modulating host gene expression. Notably, EBV ncRNAs, especially Epstein-Barr-encoded RNAs (EBERs), target and interact with less accessible regions of host chromatin to repress a subset of genes via the inhibition of RNAPII-associated chromatin loops. This process involves the cofactor nucleolin (NCL) and its RNA recognition motifs, and depletion of either NCL or EBERs alters expression of genes crucial for host infection control, immune response, and cell cycle regulation. These findings unveil a sophisticated interplay between EBV and host chromatin.
Collapse
Affiliation(s)
- Simon Zhongyuan Tian
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
| | - Yang Yang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Duo Ning
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Ting Yu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Tong Gao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Yuqing Deng
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Ke Fang
- Department of Biomedical Engineering, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Yewen Xu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Kai Jing
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Guangyu Huang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Gengzhan Chen
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Pengfei Yin
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China
| | - Yiming Li
- Department of Biomedical Engineering, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
| | - Fuxing Zeng
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
| | - Ruilin Tian
- Department of Medical Neuroscience, School of Medicine, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
- Key University Laboratory of Metabolism and Health of Guangdong, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
| | - Meizhen Zheng
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, 518055, Shenzhen, Guangdong, China.
| |
Collapse
|
32
|
van Bentum M, Klinger B, Sieber A, Naghiloo S, Zauber H, Lehmann N, Haji M, Niquet S, Mertins P, Blüthgen N, Selbach M. Spike-in enhanced phosphoproteomics uncovers synergistic signaling responses to MEK inhibition in colon cancer cells. Nat Commun 2025; 16:4884. [PMID: 40419504 PMCID: PMC12106795 DOI: 10.1038/s41467-025-59404-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 04/23/2025] [Indexed: 05/28/2025] Open
Abstract
Targeted kinase inhibitors are a cornerstone of cancer therapy, but their success is often hindered by the complexity of cellular signaling networks that can lead to resistance. Overcoming this challenge necessitates a deep understanding of cellular signaling responses. While standard global phosphoproteomics offers extensive insights, lengthy processing times, the complexity of data interpretation, and frequent omission of crucial phosphorylation sites limit its utility. Here, we combine data-independent acquisition (DIA) with spike-in of synthetic heavy stable isotope-labeled phosphopeptides to facilitate the targeted detection of particularly informative phosphorylation sites. Our spike-in enhanced detection in DIA (SPIED-DIA) approach integrates the improved sensitivity of spike-in-based targeted detection with the discovery potential of global phosphoproteomics into a simple workflow. We employed this method to investigate synergistic signaling responses in colorectal cancer cell lines following MEK inhibition. Our findings highlight that combining MEK inhibition with growth factor stimulation synergistically activates JNK signaling in HCT116 cells. This synergy emphasizes the therapeutic potential of concurrently targeting MEK and JNK pathways, as evidenced by the significantly impaired growth of HCT116 cells when treated with both inhibitors. Our results demonstrate that SPIED-DIA effectively identifies synergistic signaling responses in colorectal cancer cells, presenting a valuable tool for uncovering new therapeutic targets and strategies in cancer treatment.
Collapse
Affiliation(s)
- Mirjam van Bentum
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092, Berlin, Germany
- Institute for Theoretical Biology, Faculty of Life Sciences, Humboldt-Universität zu Berlin, Unter den Linden 6, 10099, Berlin, Germany
| | - Bertram Klinger
- Institute for Theoretical Biology, Faculty of Life Sciences, Humboldt-Universität zu Berlin, Unter den Linden 6, 10099, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Anja Sieber
- Institute for Theoretical Biology, Faculty of Life Sciences, Humboldt-Universität zu Berlin, Unter den Linden 6, 10099, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Sheyda Naghiloo
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092, Berlin, Germany
| | - Henrik Zauber
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092, Berlin, Germany
| | - Nadine Lehmann
- Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Mohamed Haji
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092, Berlin, Germany
| | - Sylvia Niquet
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Nils Blüthgen
- Institute for Theoretical Biology, Faculty of Life Sciences, Humboldt-Universität zu Berlin, Unter den Linden 6, 10099, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Matthias Selbach
- Max Delbrück Center for Molecular Medicine, Robert-Rössle-Str. 10, 13092, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
33
|
Song PY, Tsai CE, Chen YC, Huang YW, Chen PP, Wang TH, Hu CY, Chen PY, Ku C, Hsia KC, Ting SY. An interbacterial cysteine protease toxin inhibits cell growth by targeting type II DNA topoisomerases GyrB and ParE. PLoS Biol 2025; 23:e3003208. [PMID: 40424468 DOI: 10.1371/journal.pbio.3003208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Bacteria deploy a diverse arsenal of toxic effectors to antagonize competitors, profoundly influencing the composition of microbial communities. Previous studies have identified an interbacterial toxin predicted to exhibit proteolytic activity that is broadly distributed among gram-negative bacteria. However, the precise mechanism of intoxication remains unresolved. Here, we demonstrate that one such protease toxin from Escherichia coli, Cpe1, disrupts DNA replication and chromosome segregation by cleaving conserved sequences within the ATPase domain of type II DNA topoisomerases GyrB and ParE. This cleavage effectively inhibits topoisomerase-mediated relaxation of supercoiled DNA, resulting in impaired bacterial growth. Cpe1 belongs to the papain-like cysteine protease family and is associated with toxin delivery pathways, including the type VI secretion system and contact-dependent growth inhibition. The structure of Cpe1 in complex with its immunity protein reveals a neutralization mechanism involving competitive substrate binding rather than active site occlusion, distinguishing it from previously characterized effector-immunity pairs. Our findings unveil a unique mode of interbacterial intoxication and provide insights into how bacteria protect themselves from self-poisoning by protease toxins.
Collapse
Affiliation(s)
- Pin-Yi Song
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chia-En Tsai
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yung-Chih Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Yu-Wen Huang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Po-Pang Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-Haw Wang
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | - Chao-Yuan Hu
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Po-Yin Chen
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Chuan Ku
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| | - Kuo-Chiang Hsia
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - See-Yeun Ting
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
34
|
Tripathy MK, Wang H, Slocum RD, Jiang HW, Nam JC, Sabharwal T, Veerappa R, Brown KA, Cai X, Faull PA, Clark G, Roux SJ. Modified pea apyrase has altered nuclear functions and enhances the growth of yeast and Arabidopsis. FRONTIERS IN PLANT SCIENCE 2025; 16:1584871. [PMID: 40491823 PMCID: PMC12146327 DOI: 10.3389/fpls.2025.1584871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/28/2025] [Indexed: 06/11/2025]
Abstract
Apyrases (NTPDases) regulate growth and development in multiple eukaryotic organisms and function in multiple sub-cellular locales. An earlier report showed that the ectopic expression of psNTP9 (PS), a chromatin-associated pea (Pisum sativum) apyrase, enhanced the uptake of inorganic phosphate (Pi) and increased the growth of yeast and Arabidopsis. In this follow-up study, we generated a modified form of PS, abbreviated DM ("double mutant"), in which two-point mutations, S208L and P216R, were introduced into its DNA-binding domain. Ectopic expression of DM increased the growth of yeast and Arabidopsis, the seed yield of Arabidopsis, and the Pi content of yeast and Arabidopsis grown in Murashige-Skoog media beyond that effected by PS. Both the PS and DM proteins co-purified with nuclei and chromatin-associated proteins from yeast and Arabidopsis, and expression of their transgenes in these model organisms produced gene expression profiles that would be expected to promote increased growth and Pi uptake. Chromatin immunoprecipitation (ChIP)-seq analyses showed that PS and DM have largely different binding sites on yeast chromatin, including sites in promoters of numerous genes that are differentially-expressed in PS and DM transgenic lines. These results are consistent with the hypothesis that the effects of ectopically expressing the pea apyrase in yeast and in Arabidopsis are mediated, at least in part, by its activities in the nucleus that impact transcription.
Collapse
Affiliation(s)
- Manas K. Tripathy
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Huan Wang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Robert D. Slocum
- Department of Biological Sciences, Goucher College, Towson, MD, United States
| | - Han-Wei Jiang
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Ji-Chul Nam
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Tanya Sabharwal
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Roopadarshini Veerappa
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Katherine A. Brown
- The Oden Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, United States
- Cavendish Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Xingbo Cai
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Peter Allen Faull
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Greg Clark
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| | - Stanley J. Roux
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
35
|
Xu Z, Wang Y, Xie T, Luo R, Ni HL, Xiang H, Tang S, Tan S, Fang R, Ran P, Zhang Q, Xu X, Tian S, He F, Yang W, Ding C. Panoramic spatial enhanced resolution proteomics (PSERP) reveals tumor architecture and heterogeneity in gliomas. J Hematol Oncol 2025; 18:58. [PMID: 40420200 DOI: 10.1186/s13045-025-01710-5] [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: 09/04/2024] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
The spatial proteomic profiling of complex tissues is essential for investigating cellular function in physiological and pathological states. However, the imbalance among resolution, protein coverage, and expense precludes their systematic application to analyze whole tissue sections in an unbiased manner and with high resolution. Here, we introduce panoramic spatial enhanced resolution proteomics (PSERP), a method that combines tissue expansion, automated sample segmentation, and tryptic digestion with high-throughput proteomic profiling. The PSERP approach facilitates rapid quantitative profiling of proteomic spatial variability in whole tissue sections at sub-millimeter resolution. We demonstrated the utility of this method for determining the streamlined large-scale spatial proteomic features of gliomas. Specifically, we profiled spatial proteomic features for nine glioma samples across three different mutation types (IDH1-WT/EGFR-mutant, IDH1-mutant, and IDH1/EGFR-double-WT gliomas) at sub-millimeter resolution (corresponding to a total of 2,230 voxels). The results revealed over 10,000 proteins identified in a single slide, which helps us to portray the diverse proteins and pathways with spatial abundance patterns in the context of tumor heterogeneity and cellular features. Our spatial proteomic data revealed distinctive proteomic features of malignant and non-malignant tumor regions and depicted the distribution of proteins from tumor centers to tumor borders and non-malignant tumor regions. Through integrative analysis with single-cell transcriptomic data, we elucidated the cellular composition and cell-cell communications in a spatial context. Our PSERP also includes a spatially resolved tumor-specific peptidome identification workflow that not only enables us to elucidate the spatial expression patterns of tumor-specific peptides in glioma samples with different genomic types but also provides us with opportunities to select combinations of tumor-specific mutational peptides whose expression could cover the maximum tumor regions for future immune therapies. We further demonstrated that combining tumor-specific peptides might enhance the efficacy of immunotherapy in both patient-derived cell (PDC) and patient-derived xenograft (PDX) models. PSERP efficiently retains precise spatial proteomic information within the tissue context and provides a deeper understanding of tissue biology and pathology at the molecular level.
Collapse
Affiliation(s)
- Ziyan Xu
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yunzhi Wang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Tao Xie
- Department of Neurosurgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Heng-Li Ni
- Department of Pathology, Children's Hospital of Soochow University, Soochow University, Suzhou, JiangSu Province, 215000, The People's Republic of China
| | - Hang Xiang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shaoshuai Tang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Subei Tan
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Rundong Fang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Peng Ran
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qiao Zhang
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiaomeng Xu
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Sha Tian
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fuchu He
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Beijing, China
- Research Unit of Proteomics Driven Cancer Precision Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenjun Yang
- Department of Pediatric Orthopedics, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China.
| | - Chen Ding
- Clinical Research Center for Cell-based Immunotherapy of Shanghai Pudong Hospital, Fudan University Pudong Medical Center, State Key Laboratory of Genetics and Development of Complex Phenotypes, School of Life Sciences, Human Phenome Institute, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- Departments of Cancer Research Institute, Affiliated Cancer Hospital of Xinjiang Medical University, Xinjiang Key Laboratory of Translational Biomedical Engineering, Urumqi 830000, China.
| |
Collapse
|
36
|
Marotta G, Osti D, Zaccheroni E, Costanza B, Faletti S, Marinaro A, Richichi C, Mesa D, Rodighiero S, Soriani C, Migliaccio E, Ruscitto F, Priami C, Sigismund S, Manetti F, Polli D, Beznusenko GV, Rusu MC, Favero F, Corà D, Silvestris DA, Gallo A, Gambino V, Alfieri F, Gandini S, Schmitt MJ, Gargiulo G, Noberini R, Bonaldi T, Pelicci G. Metabolic traits shape responses to LSD1-directed therapy in glioblastoma tumor-initiating cells. SCIENCE ADVANCES 2025; 11:eadt2724. [PMID: 40408499 DOI: 10.1126/sciadv.adt2724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 04/17/2025] [Indexed: 05/25/2025]
Abstract
Lysine-specific histone demethylase 1A (LSD1) is an epigenetic regulator involved in various biological processes, including metabolic pathways. We demonstrated the therapeutic potential of its pharmacological inhibition in glioblastoma using DDP_38003 (LSD1i), which selectively targets tumor-initiating cells (TICs) by hampering their adaptability to stress. Through biological, metabolic, and omic approaches, we now show that LSD1i acts as an endoplasmic reticulum (ER) stressor, activating the integrated stress response and altering mitochondrial structure and function. These effects impair TICs' oxidative metabolism and generate reactive oxygen species, further amplifying cellular stress. LSD1i also impairs TICs' glycolytic activity, causing their metabolic decline. TICs with enhanced glycolysis benefit from LSD1-directed therapy. Conversely, metabolically silent TICs mantain ER and mitochondrial homeostasis, adapting to stress conditions, including LSD1i treatment. A dropout short hairpin RNA screening identifies postglycosylphosphatidylinositol attachment to proteins inositol deacylase 1 (PGAP1) as a mediator of resistance to LSD1i. Disruptions in ER and mitochondrial balance holds promise for improving LSD1-targeted therapy efficacy and overcoming treatment resistance.
Collapse
Affiliation(s)
- Giulia Marotta
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Daniela Osti
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Elena Zaccheroni
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Brunella Costanza
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Stefania Faletti
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
- Human Technopole, Viale Rita Levi-Montalcini, 1, Milan 20157, Italy
| | - Adriana Marinaro
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Cristina Richichi
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Deborah Mesa
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Simona Rodighiero
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Chiara Soriani
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Enrica Migliaccio
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Federica Ruscitto
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Chiara Priami
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Sara Sigismund
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan Italy
| | | | - Dario Polli
- Department of Physics, Politecnico di Milano, Milan, Italy
- CNR Institute for Photonics and Nanotechnology (CNR-IFN), Milan, Italy
| | | | - Mara-Camelia Rusu
- Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Francesco Favero
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, Novara 28100, Italy
| | - Davide Corà
- Department of Translational Medicine, Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, Novara 28100, Italy
| | - Domenico A Silvestris
- Unit of Genetics and Epigenetic of Pediatric Cancer, Oncohaematology Department, IRCCS Ospedale Pediatrico Bambino Gesù, Viale di San Paolo 15, Rome 00146, Italy
| | - Angela Gallo
- Unit of Genetics and Epigenetic of Pediatric Cancer, Oncohaematology Department, IRCCS Ospedale Pediatrico Bambino Gesù, Viale di San Paolo 15, Rome 00146, Italy
| | - Valentina Gambino
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Fabio Alfieri
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Sara Gandini
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Matthias J Schmitt
- Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Gaetano Gargiulo
- Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Roberta Noberini
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
| | - Tiziana Bonaldi
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
- Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan Italy
| | - Giuliana Pelicci
- Department of Experimental Oncology, European Institute of Oncology (IEO), IRCCS, Milan 20139, Italy
- Department of Translational Medicine, University of Piemonte Orientale, Novara 28100, Italy
| |
Collapse
|
37
|
Abdullah HQ, Levanon NL, Perach M, Grupper M, Ziv T, Lewinson O. When less is more: Counterintuitive stoichiometries and cellular abundances are essential for ABC transporters' function. SCIENCE ADVANCES 2025; 11:eadq7470. [PMID: 40397753 PMCID: PMC12094219 DOI: 10.1126/sciadv.adq7470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 04/16/2025] [Indexed: 05/23/2025]
Abstract
Prokaryotes acquire essential nutrients primarily through adenosine triphosphate-binding cassette (ABC) importers, consisting of an adenosine triphosphatase, a permease, and a substrate-binding protein. These importers are highly underrepresented in proteomic databases, limiting our knowledge about their cellular copy numbers, component stoichiometry, and the mechanistic implications of these parameters. We developed a tailored proteomic approach to compile the most comprehensive dataset to date of the Escherichia coli "ABC importome." Functional assays and analyses of deletion strains revealed mechanistic features linking molecular mechanisms to cellular abundances, colocalization, and component stoichiometries. We observed four to five orders of magnitude variation in import system abundances, with copy numbers tuned to nutrient hierarchies essential for growth. Abundances of substrate-binding proteins are unrelated to their substrate binding affinities but are tightly yet inversely correlated with their interaction affinity with permeases. Counterintuitive component stoichiometries are crucial for function, offering insights into the design principles of multicomponent protein systems, potentially extending beyond ABC importers.
Collapse
Affiliation(s)
- Hiba Qasem Abdullah
- Department of Molecular Microbiology, Bruce and Ruth Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nurit Livnat Levanon
- Department of Molecular Microbiology, Bruce and Ruth Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Michal Perach
- Department of Molecular Microbiology, Bruce and Ruth Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Moti Grupper
- Infectious Disease Unit, Rambam Health Care Campus, Haifa, Israel
| | - Tamar Ziv
- Smoler Proteomics Center, Technion-Israel Institute of Technology, Haifa, Israel
| | - Oded Lewinson
- Department of Molecular Microbiology, Bruce and Ruth Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
38
|
Fu Y, Trautwein-Schult A, Piersma S, Sun C, Westra J, de Jong A, Becher D, van Dijl JM. Characterization of outer membrane vesicles of Aggregatibacter actinomycetemcomitans serotypes a, b and c and their interactions with human neutrophils. Int J Med Microbiol 2025; 319:151655. [PMID: 40424897 DOI: 10.1016/j.ijmm.2025.151655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 05/02/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025] Open
Abstract
Aggregatibacter actinomycetemcomitans (Aa) is a Gram-negative oral pathogen associated with periodontitis and systemic diseases. Seven serotypes of Aa are known, with serotypes a, b and c being most prevalent worldwide. Interestingly, serotype a, b and c isolates present differences in virulence. This focuses interest on their secreted virulence factors. Gram-negative bacteria evolved a specific protein secretion mechanism, based on the release of outer membrane vesicles (OMVs) with a protein cargo. The present study was therefore aimed at investigating whether differences in the protein cargo of OMVs could be associated with the differential virulence of Aa serotypes a, b or c. Accordingly, the different OMV proteomes were defined by mass spectrometry and infection assays were performed with human neutrophils that represent the main innate defense against oral pathogens like Aa. Subsequently, we correlated the OMV proteome data with the observed OMV-neutrophil interactions. A total of 276 OMV-associated proteins was identified, including 53 known virulence factors. Interestingly, OMVs from Aa isolates with different serotypes displayed similar protein cargo, but the relative quantities differed. OMVs of serotype a isolates were exceptional in carrying CRISPR proteins with a potential role in virulence. Intriguingly, Aa OMVs mostly coated the neutrophil surface, triggering formation of neutrophil extracellular traps (NETs). Conversely, the NETs captured Aa OMVs. Since the observed OMV-neutrophil interplay will occur at a distance from the OMV-producing bacteria, we postulate that it allows the bacteria to evade capture and elimination by neutrophils.
Collapse
Affiliation(s)
- Yanyan Fu
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands
| | - Anke Trautwein-Schult
- University of Greifswald, Institute of Microbiology, Department of Microbial Proteomics, Greifswald, Germany
| | - Sjouke Piersma
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands
| | - Chang Sun
- University of Groningen, University Medical Center Groningen, Department of Biomedical Sciences of cells and Systems, Groningen, the Netherlands
| | - Johanna Westra
- University of Groningen, University Medical Center Groningen, Department of Rheumatology and Clinical Immunology, Groningen, the Netherlands
| | - Anne de Jong
- University of Groningen, Groningen Biomolecular Sciences and Biotechnology Institute, Department of Molecular Genetics, Groningen, the Netherlands
| | - Dörte Becher
- University of Greifswald, Institute of Microbiology, Department of Microbial Proteomics, Greifswald, Germany
| | - Jan Maarten van Dijl
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology, Groningen, the Netherlands.
| |
Collapse
|
39
|
Kim G, Grams RJ, Hsu KL. Advancing Covalent Ligand and Drug Discovery beyond Cysteine. Chem Rev 2025. [PMID: 40404146 DOI: 10.1021/acs.chemrev.5c00001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
Targeting intractable proteins remains a key challenge in drug discovery, as these proteins often lack well-defined binding pockets or possess shallow surfaces not readily addressed by traditional drug design. Covalent chemistry has emerged as a powerful solution for accessing protein sites in difficult to ligand regions. By leveraging activity-based protein profiling (ABPP) and LC-MS/MS technologies, academic groups and industry have identified cysteine-reactive ligands that enable selective targeting of challenging protein sites to modulate previously inaccessible biological pathways. Cysteines within a protein are rare, however, and developing covalent ligands that target additional residues hold great promise for further expanding the ligandable proteome. This review highlights recent advancements in targeting amino acids beyond cysteine binding with an emphasis on tyrosine- and lysine-directed covalent ligands and their applications in chemical biology and therapeutic development. We outline the process of developing covalent ligands using chemical proteomic methodology, highlighting recent successful examples and discuss considerations for future expansion to additional amino acid sites on proteins.
Collapse
Affiliation(s)
- Gibae Kim
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - R Justin Grams
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| | - Ku-Lung Hsu
- Department of Chemistry, University of Texas at Austin, Austin, Texas 78712, United States
| |
Collapse
|
40
|
Devanathan SK, Li YR, Shelton SB, Nguyen J, Tseng WC, Shah NM, Mercado M, Miller KM, Xhemalçe B. MePCE promotes homologous recombination through coordinating R-loop resolution at DNA double-stranded breaks. Cell Rep 2025; 44:115740. [PMID: 40411785 DOI: 10.1016/j.celrep.2025.115740] [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: 08/17/2024] [Revised: 02/26/2025] [Accepted: 05/05/2025] [Indexed: 05/26/2025] Open
Abstract
MePCE is a multifunctional protein that regulates the positive transcription elongation factor b (P-TEFb) partitioning between the nucleosol and chromatin. MePCE's role in sequestering P-TEFb in the nucleosol via the 7SK ribonuclear protein complex (RNPc) is clear, but its functions on chromatin remain obscure. We report that chromatin-associated MePCE interacts with R-loop processing and DNA repair factors. MePCE is recruited to DNA double-stranded breaks (DSBs), and MePCE depletion impairs DSB repair by homologous recombination (HR), decreases RAD51 loading, and enhances R-loop levels at AsiSI-induced DSBs at specific genomic locations. Besides decreasing specific R-loop processing factors and chromatin remodelers, MePCE depletion increases the interaction with R-loops of the other constitutive member of the 7SK RNPc, LARP7, which is degraded by BRCA1/BARD1 upon DSB. Overall, our results uncover dynamic regulation of the 7SK RNPc at DSBs during the DSB repair process and explain the recently observed synthetic lethality of MePCE and BRCA1 deficiency.
Collapse
Affiliation(s)
- Sravan K Devanathan
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
| | - Yi-Ru Li
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA; Department of Biochemistry and Winship Cancer Center, Emory University School of Medicine, 1510 Clifton Road NE, Atlanta, GA 30322, USA
| | - Samantha B Shelton
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
| | - Joshuah Nguyen
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA; Department of Biochemistry and Winship Cancer Center, Emory University School of Medicine, 1510 Clifton Road NE, Atlanta, GA 30322, USA
| | - Wei-Che Tseng
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA; Department of Radiation Oncology and Winship Cancer Center, Emory University School of Medicine, 1750 Haygood Drive NE, Atlanta, GA 30307, USA
| | - Nakul M Shah
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
| | - Marvin Mercado
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA
| | - Kyle M Miller
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA; Department of Radiation Oncology and Winship Cancer Center, Emory University School of Medicine, 1750 Haygood Drive NE, Atlanta, GA 30307, USA
| | - Blerta Xhemalçe
- Department of Molecular Biosciences, University of Texas at Austin, 2500 Speedway, Austin, TX 78712, USA; Department of Biochemistry and Winship Cancer Center, Emory University School of Medicine, 1510 Clifton Road NE, Atlanta, GA 30322, USA.
| |
Collapse
|
41
|
Wilcken S, Koutsandrea PH, Bakker T, Kulik A, Orthwein T, Franz-Wachtel M, Harbig T, Nieselt KK, Forchhammer K, Brötz-Oesterhelt H, Macek B, Mordhorst S, Kaysser L, Gust B. The TetR-like regulator Sco4385 and Crp-like regulator Sco3571 modulate heterologous production of antibiotics in Streptomyces coelicolor M512. Appl Environ Microbiol 2025; 91:e0231524. [PMID: 40183567 DOI: 10.1128/aem.02315-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/09/2025] [Indexed: 04/05/2025] Open
Abstract
Heterologous expression in well-studied model strains is a routinely applied method to investigate biosynthetic pathways. Here, we pursue a comparative approach of large-scale DNA-affinity-capturing assays (DACAs) coupled with semi-quantitative mass spectrometry (MS) to identify putative regulatory proteins from Streptomyces coelicolor M512, which bind to the heterologously expressed biosynthetic gene clusters (BGCs) of the liponucleoside antibiotics caprazamycin and liposidomycin. Both gene clusters share an almost identical genetic arrangement, including the location of promoter regions, as detected by RNA sequencing. A total of 2,214 proteins were trapped at the predicted promoter regions, with only three binding to corresponding promoters in both gene clusters. Among these, the overexpression of a yet uncharacterized TetR-family regulator (TFR), Sco4385, increased caprazamycin but not liposidomycin production. Protein-DNA interaction experiments using biolayer interferometry confirmed the binding of Sco4385 to Pcpz10 and PlpmH at different locations within both promoter regions, which might explain its functional variance. Sequence alignment allowed the determination of a consensus sequence present in both promoter regions, to which Sco4385 was experimentally shown to bind. Furthermore, we found that the overexpression of the Crp regulator, Sco3571, leads to a threefold increase in caprazamycin and liposidomycin production yields, possibly due to an increased expression of a precursor pathway.IMPORTANCEStreptomycetes are well-studied model organisms for the biosynthesis of pharmaceutically, industrially, and biotechnologically valuable metabolites. Their naturally broad repertoire of natural products can be further exploited by heterologous expression of biosynthetic gene clusters (BGCs) in non-native host strains. This approach forces the host to adapt to a new regulatory and metabolic environment. In our study, we demonstrate that a host regulator not only interacts with newly incorporated gene clusters but also regulates precursor supply for the produced compounds. We present a comprehensive study of regulatory proteins that interact with two genetically similar gene clusters for the biosynthesis of liponucleoside antibiotics. Thereby, we identified regulators of the heterologous host that influence the production of the corresponding antibiotic. Surprisingly, the regulatory interaction is highly specific for each biosynthetic gene cluster, even though they encode largely structurally similar metabolites.
Collapse
Affiliation(s)
- Sarah Wilcken
- Pharmaceutical Biology, Pharmaceutical Institute, Eberhard-Karls-University Tübingen, Tübingen, Germany
- Partner Site Tübingen, German Centre for Infection Research (DZIF), Tübingen, Germany
| | | | - Tomke Bakker
- Pharmaceutical Biology, Pharmaceutical Institute, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Andreas Kulik
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Tim Orthwein
- Department of Microbiology and Organismic Interactions, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Mirita Franz-Wachtel
- Proteome Center Tübingen, Institute of Cell Biology, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Theresa Harbig
- Interfaculty Institute for Bioinformatics and Medical Informatics, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Kay Katja Nieselt
- Interfaculty Institute for Bioinformatics and Medical Informatics, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Karl Forchhammer
- Department of Microbiology and Organismic Interactions, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Heike Brötz-Oesterhelt
- Partner Site Tübingen, German Centre for Infection Research (DZIF), Tübingen, Germany
- Department of Microbial Bioactive Compounds, Interfaculty Institute of Microbiology and Infection Medicine, Eberhard-Karls-University Tübingen, Tübingen, Germany
- Cluster of Excellence Controlling Microbes to Fight Infections, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Boris Macek
- Proteome Center Tübingen, Institute of Cell Biology, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Silja Mordhorst
- Pharmaceutical Biology, Pharmaceutical Institute, Eberhard-Karls-University Tübingen, Tübingen, Germany
| | - Leonard Kaysser
- Institute for Drug Discovery, Department of Pharmaceutical Biology, Leipzig University, Leipzig, Germany
| | - Bertolt Gust
- Pharmaceutical Biology, Pharmaceutical Institute, Eberhard-Karls-University Tübingen, Tübingen, Germany
- Partner Site Tübingen, German Centre for Infection Research (DZIF), Tübingen, Germany
| |
Collapse
|
42
|
Fischer P, Thoms M, Lau B, Denk T, Kuvshinova M, Berninghausen O, Flemming D, Hurt E, Beckmann R. H/ACA snR30 snoRNP guides independent 18S rRNA subdomain formation. Nat Commun 2025; 16:4720. [PMID: 40399280 PMCID: PMC12095548 DOI: 10.1038/s41467-025-59656-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 04/25/2025] [Indexed: 05/23/2025] Open
Abstract
Ribosome biogenesis follows a cascade of pre-rRNA folding and processing steps, coordinated with ribosomal protein incorporation. Nucleolar 90S pre-ribosomes are well-described stable intermediates, composed of pre-18S rRNA, ribosomal S-proteins, U3 snoRNA, and ~70 assembly factors. However, how numerous snoRNAs control pre-rRNA modification and folding during early maturation events remains unclear. We identify snR30 (human U17), the only essential H/ACA snoRNA in yeast, which binds with Cbf5-Gar1-Nop10-Nhp2 to a pre-18S rRNA subdomain containing platform helices and ES6 of the 40S central domain. Integration into the 90S is blocked by RNA hybridization with snR30. The snoRNP complex coordinates the recruitment of early assembly factors Krr1-Utp23-Kri1 and ribosomal proteins uS11-uS15, enabling isolated subdomain assembly. Krr1-dependent release of snR30 culminates in integration of the platform into the 90S. Our study reveals the essential role of snR30 in chaperoning central domain formation as a discrete assembly unit externalized from the pre-ribosomal core.
Collapse
MESH Headings
- RNA, Ribosomal, 18S/metabolism
- RNA, Ribosomal, 18S/genetics
- RNA, Ribosomal, 18S/chemistry
- Saccharomyces cerevisiae Proteins/metabolism
- Saccharomyces cerevisiae Proteins/genetics
- Saccharomyces cerevisiae/metabolism
- Saccharomyces cerevisiae/genetics
- Humans
- RNA, Small Nucleolar/metabolism
- RNA, Small Nucleolar/genetics
- Ribonucleoproteins, Small Nucleolar/metabolism
- Ribonucleoproteins, Small Nucleolar/genetics
- RNA Precursors/metabolism
- RNA Precursors/genetics
- Ribosomal Proteins/metabolism
- Ribosomes/metabolism
- Protein Binding
Collapse
Affiliation(s)
- Paulina Fischer
- Biochemistry Center, Heidelberg University, Heidelberg, Germany
| | - Matthias Thoms
- Department of Biochemistry, Gene Center, University of Munich, Munich, Germany
| | - Benjamin Lau
- Biochemistry Center, Heidelberg University, Heidelberg, Germany
- Molecular Systems Biology Unit, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, Heidelberg, Germany
| | - Timo Denk
- Department of Biochemistry, Gene Center, University of Munich, Munich, Germany
| | | | - Otto Berninghausen
- Department of Biochemistry, Gene Center, University of Munich, Munich, Germany
| | - Dirk Flemming
- Biochemistry Center, Heidelberg University, Heidelberg, Germany
| | - Ed Hurt
- Biochemistry Center, Heidelberg University, Heidelberg, Germany.
| | - Roland Beckmann
- Department of Biochemistry, Gene Center, University of Munich, Munich, Germany.
| |
Collapse
|
43
|
Wolfgramm H, Saade C, Harms M, Busch LM, Lange J, Schedlowski M, Surmann K, Gesell Salazar M, Hentschker C, Steil L, Michalik S, Völker U, Reder A. pTripleTREP - A vector for tightly controlled expression and purification of virulence factors in Staphylococcus aureus. Microb Cell Fact 2025; 24:115. [PMID: 40394585 DOI: 10.1186/s12934-025-02736-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Recombinant proteins facilitate and contribute to detailed studies of the virulence mechanisms and pathophysiology of the major human pathogen Staphylococcus aureus. Of particular interest are secreted virulence factors. However, due to their potential toxicity and specific post-translational processing, virulence factors are difficult targets for heterologous protein production. Purified proteins with native conformation and adequate purity can therefore often only be achieved by elaborate multi-step purification workflows. While homologous expression in S. aureus theoretically offers a promising alternative in this regard, its application remains limited due to the lack of systems that ensure both tightly controlled expression and subsequent efficient purification. RESULTS To bridge this gap, we present pTripleTREP as a versatile expression vector for S. aureus, which enables the homologous expression and purification of staphylococcal virulence factors. It features a strong SigA-dependent staphylococcal promoter overlapped by three tetracycline responsive elements (TRE), which ensures tight repression under control conditions and high expression levels upon induction of the target gene. This allowed very precise controlled production of the exemplary targets, serine protease-like protein A (SplA) and B (SplB). A simple single-step protein purification workflow using a Twin-Strep-tag and Strep-Tactin®XT coated magnetic beads yielded endotoxin-free Spl samples with purities above 99%. Thereby, the homologous production host facilitates native secretion and maturation without the need to engineer the target gene sequence. Proper signal peptide cleavage and the corresponding enzymatic activity of the generated protein products were confirmed for SplA and B. CONCLUSION The expression vector pTripleTREP adds an important element to the staphylococcal molecular toolbox, facilitating the tightly controlled homologous expression and rapid native purification of secreted staphylococcal virulence factors. The optimised architecture and genetic features of the vector additionally provide a solid background for further applications such as plasmid-based complementation or interaction studies. Thus, pTripleTREP will support research on the role of staphylococcal virulence factors, paving the way for future therapeutic strategies to combat this pathogen.
Collapse
Affiliation(s)
- Hannes Wolfgramm
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Christopher Saade
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Marco Harms
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Larissa M Busch
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Josephine Lange
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Maximilian Schedlowski
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Kristin Surmann
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Manuela Gesell Salazar
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Christian Hentschker
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Leif Steil
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stephan Michalik
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany.
| | - Alexander Reder
- Interfaculty Institute of Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
44
|
Merold V, Bekere I, Kretschmer S, Schnell AF, Kmiec D, Sivarajan R, Lammens K, Liu R, Mergner J, Teppert J, Hirschenberger M, Henrici A, Hammes S, Buder K, Weitz M, Hackmann K, Koenig LM, Pichlmair A, Schwierz N, Sparrer KMJ, Lee-Kirsch MA, de Oliveira Mann CC. Structural basis for OAS2 regulation and its antiviral function. Mol Cell 2025:S1097-2765(25)00406-X. [PMID: 40412389 DOI: 10.1016/j.molcel.2025.05.001] [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: 10/11/2024] [Revised: 02/01/2025] [Accepted: 05/01/2025] [Indexed: 05/27/2025]
Abstract
Oligoadenylate synthetase (OAS) proteins are immune sensors for double-stranded RNA and are critical for restricting viruses. OAS2 comprises two OAS domains, only one of which can synthesize 2'-5'-oligoadenylates for RNase L activation. Existing structures of OAS1 provide a model for enzyme activation, but they do not explain how multiple OAS domains discriminate RNA length. Here, we discover that human OAS2 exists in an auto-inhibited state as a zinc-mediated dimer and present a mechanism for RNA length discrimination: the catalytically deficient domain acts as a molecular ruler that prevents autoreactivity to short RNAs. We demonstrate that dimerization and myristoylation localize OAS2 to Golgi membranes and that this is required for OAS2 activation and the restriction of viruses that exploit the endomembrane system for replication, e.g., coronaviruses. Finally, our results highlight the non-redundant role of OAS proteins and emphasize the clinical relevance of OAS2 by identifying a patient with a loss-of-function mutation associated with autoimmune disease.
Collapse
Affiliation(s)
- Veronika Merold
- Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| | - Indra Bekere
- Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| | - Stefanie Kretschmer
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Adrian F Schnell
- Institute of Physics, University of Augsburg, Augsburg 86159, Germany
| | - Dorota Kmiec
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Rinu Sivarajan
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany
| | - Katja Lammens
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Rou Liu
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany
| | - Julia Mergner
- Bavarian Center for Biomolecular Mass Spectrometry at Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Munich 81675, Germany
| | - Julia Teppert
- Division of Clinical Pharmacology, University Hospital, Ludwig-Maximilians-Universität München, Munich 80337, Germany
| | | | - Alexander Henrici
- School of Medicine, Institute of Virology, Technical University of Munich, Munich 81675, Germany
| | - Sarah Hammes
- Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Garching 85748, Germany
| | - Kathrin Buder
- University Hospital Tuebingen, University Children's Hospital, Department of General Pediatrics and Hematology/Oncology, Tuebingen 72076, Germany
| | - Marcus Weitz
- University Hospital Tuebingen, University Children's Hospital, Department of General Pediatrics and Hematology/Oncology, Tuebingen 72076, Germany
| | - Karl Hackmann
- Institute for Clinical Genetics, University Hospital Carl Gustav Carus at TUD Dresden University of Technology, Dresden 01307, Germany
| | - Lars M Koenig
- Division of Clinical Pharmacology, University Hospital, Ludwig-Maximilians-Universität München, Munich 80337, Germany
| | - Andreas Pichlmair
- School of Medicine, Institute of Virology, Technical University of Munich, Munich 81675, Germany; Helmholtz Center Munich, Systems Virology, Neuherberg 85764, Germany; German Center for Infection Research, Partner site Munich, Munich 81675, Germany
| | - Nadine Schwierz
- Institute of Physics, University of Augsburg, Augsburg 86159, Germany
| | - Konstantin M J Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Ulm 89081, Germany; German Center for Neurodegenerative Diseases, Ulm 89081, Germany
| | - Min Ae Lee-Kirsch
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; University Center for Rare Diseases, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden 01307, Germany; German Center for Child and Adolescent Health, partner site Leipzig/Dresden, Dresden 01307, Germany
| | - Carina C de Oliveira Mann
- Department of Bioscience, TUM School of Natural Sciences, Technical University of Munich, Garching 85748, Germany.
| |
Collapse
|
45
|
Wang A, Bolnick DI. Among-population proteomic differences in Schistocephalus solidus based on excretory/secretory and total body protein predictions. Parasit Vectors 2025; 18:180. [PMID: 40394694 DOI: 10.1186/s13071-025-06807-x] [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: 10/14/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Parasites secrete and excrete a variety of molecules that evolved to help establish and sustain infections within hosts. Parasite adaptation to their host may lead to between-population divergence in these excretory and secretory products (ESPs), but few studies have tested for intraspecific variation in helminth proteomes. METHODS Schistocephalus solidus is a cestode that parasitizes the threespine stickleback, Gasterosteus aculeatus. We used an ultra-performance liquid chromatography-mass spectrometry protocol to characterize the ESPs and whole-body proteome of S. solidus. Specifically, we characterized the proteome of S. solidus at the plerocercoid stage from wild-caught stickleback from three lakes on Vancouver Island (British Columbia, Canada) and one lake in Alaska (USA). We tested for differences in proteome composition among the four populations and specifically between ESPs and body tissue. RESULTS Overall, we identified 1362 proteins in the total proteome of S. solidus, with 542 of the 1362 proteins detected exclusively in the ESPs. Of the ESP proteins, we found signaling peptides and transmembrane proteins that had not been previously detected or characterized in S. solidus. We also found that protein spectrum counts varied greatly among all lake populations. CONCLUSIONS These population-level differences were observed in both ESP and whole-body tissue types. Our study suggests that S. solidus can excrete and secrete a wide range of proteins which are distinct among populations. These differences might reflect plastic responses to host genotype differences, or evolved adaptations by Schistocephalus to different local host populations.
Collapse
Affiliation(s)
- Anni Wang
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT, 06269, USA
| | - Daniel I Bolnick
- Department of Ecology and Evolutionary Biology, University of Connecticut, Storrs, CT, 06269, USA.
| |
Collapse
|
46
|
Martinez-Castillo A, Barriales D, Azkargorta M, Zalamea JD, Ardá A, Jimenez-Barbero J, Gonzalez-Lopez M, Aransay AM, Marín-López A, Fikrig E, Elortza F, Anguita J, Abrescia NGA. Structural and functional significance of Aedes aegypti AgBR1 flavivirus immunomodulator. J Virol 2025; 99:e0187824. [PMID: 40272158 PMCID: PMC12090808 DOI: 10.1128/jvi.01878-24] [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: 10/23/2024] [Accepted: 03/06/2025] [Indexed: 04/25/2025] Open
Abstract
Zika virus (ZIKV), an arbovirus, relies on mosquitoes as vectors for its transmission. During blood feeding, mosquitoes inoculate saliva containing various proteins. Recently, AgBR1, an Aedes aegypti salivary gland protein, has gained attention for its immunomodulatory potential, along with another protein, called NeSt1. We have determined the crystal structure of AgBR1 at 1.2 Å resolution. Despite its chitinase-like fold, we demonstrated that AgBR1 does not bind to chitobiose or chitinhexaose, while a key mutation in the catalytic site abrogates enzymatic activity, suggesting that the protein's function has been repurposed. Our study also shows that AgBR1 and NeSt1, when presented to murine primary macrophages, alter cellular pathways related to virus entry by endocytosis, immune response, and cell proliferation. AgBR1 (and NeSt1) do not directly bind to the Zika virus or modulate its replication. We propose that their immunomodulatory effects on Zika virus transmission are through regulation of host-cell response, a consequence of evolutionary cross talk and virus opportunism. These structural and functional insights are prerequisites for developing strategies to halt the spread of mosquito-borne disease.IMPORTANCEOur study informs on the structural and functional significance of a mosquito salivary gland protein, AgBR1 (along with another protein called NeSt1), in the transmission of the Zika virus (ZIKV), a mosquito-borne virus that has caused global health concerns. By analyzing AgBR1's three-dimensional structure in combination with cellular and interaction studies, we discovered that AgBR1 does not function like typical proteins in its family-it does not degrade sugars. However, we show that it primes immune cells in a way that could help the virus enter cells more easily but not by interacting with the virus or altering viral replication. This finding is significant because it reveals how mosquito proteins, repurposed by evolution, can influence virus transmission without the virus's direct presence. Understanding how proteins like AgBR1 work could guide the development of new strategies to prevent Zika virus spread, with potential relevance for other mosquito-borne viruses.
Collapse
Affiliation(s)
- Ane Martinez-Castillo
- Structure and Cell Biology of Viruses Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE) - Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Diego Barriales
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE - BRTA, Derio, Spain
| | | | - Juan Diego Zalamea
- Structure and Cell Biology of Viruses Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE) - Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Ana Ardá
- Chemical Glycobiology Laboratory, CIC bioGUNE - BRTA, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Jesus Jimenez-Barbero
- Chemical Glycobiology Laboratory, CIC bioGUNE - BRTA, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | | | - Ana M. Aransay
- Genome Analysis Platform, CIC bioGUNE - BRTA, Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandro Marín-López
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Felix Elortza
- Proteomics Platform, CIC bioGUNE - BRTA, Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan Anguita
- Inflammation and Macrophage Plasticity Laboratory, CIC bioGUNE - BRTA, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Nicola G. A. Abrescia
- Structure and Cell Biology of Viruses Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE) - Basque Research and Technology Alliance (BRTA), Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
47
|
Li Q, Wang L, Grubb LE, Talasila M, Rodriguez Gallo MC, Mehta D, Scandola S, Uhrig RG. B4 Raf-like MAPKKK RAF24 regulates Arabidopsis thaliana flowering time through HISTONE MONO-UBIQUITINATION 2. THE NEW PHYTOLOGIST 2025. [PMID: 40394941 DOI: 10.1111/nph.70192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 04/02/2025] [Indexed: 05/22/2025]
Abstract
The timing of flowering is a critical agronomic trait governed by an extensive and sophisticated regulatory network. To date, limited understanding of how posttranslational modifications regulate flowering time exists. Here, using Arabidopsis, we resolve a role for the B4 Raf-like MAPKKK protein kinase RAF24 in regulating flowering time. Loss of RAPIDLY ACCELERATED FIBROSARCOMA24 (RAF24) led to premature flowering time through altered expression of FLC and FT. Comparative phosphoproteomic analysis of raf24 and wild-type plants revealed a list of known flowering-related phosphoproteins from distinct flowering pathways displaying downregulated phosphorylation. Of these, the RING-type ubiquitin ligase HISTONE MONO-UBIQUITINATION 2 (HUB2) lacked phosphorylation in the absence of RAF24. Absence of RAF24 induced H2Bub1 overaccumulation, with protein-protein interactome analysis of HUB2 in the presence and absence of RAF24 influencing HUB2 protein interaction partners, such as H2B. HUB2 was also found to physically interact with SUCROSE NONFERMENTING KINASE 2.4 (SnRK2.4) and SnRK2.6, known substrates of RAF24. Using phospho-mimetic and phospho-ablative plant lines, we then validated the importance of HUB2 phosphorylation at serine 314 (S314) in maintaining appropriate flowering time. Our findings uncovered a novel biological role of RAF24, as a higher-order flowering regulator, while further implicating HUB2 as a centerpiece of flowering time regulation.
Collapse
Affiliation(s)
- Qiaomu Li
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | - Le Wang
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | - Lauren E Grubb
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | - Mohana Talasila
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | | | - Devang Mehta
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
- Department of Biosystems, Katholieke Universiteit Leuven, B-3001, Leuven, Belgium
| | - Sabine Scandola
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
| | - Richard Glen Uhrig
- Department of Biological Sciences, University of Alberta, Edmonton, AB, T6G 2E9, Canada
- Department of Biochemistry, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| |
Collapse
|
48
|
Schaller E, Lamer S, Schlosser A, Stigloher C, Maher P, Decker M. Affinity-Based Protein Profiling Reveals IDH2 as a Mitochondrial Target of Cannabinol in Receptor-Independent Neuroprotection. Chemistry 2025:e202501143. [PMID: 40388656 DOI: 10.1002/chem.202501143] [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: 03/21/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/21/2025]
Abstract
Phytocannabinoids are attracting growing attention because of their potential for treatment of neurodegenerative diseases. Among them, the "minor" cannabinoid, cannabinol (CBN), has emerged as a promising neuroprotective agent, acting independently of classical cannabinoid receptors through as-yet unidentified mitochondrial targets. To uncover the molecular basis of its neuroprotective effects, we designed and synthesized a chemical probe based on CBN, incorporating a minimalist diazirine linker. Functional assays confirmed that the probe retains CBN's mitochondrial activity and exhibits strong mitochondrial enrichment, as demonstrated by fluorescence microscopy and click-correlative light and electron microscopy (click-CLEM). By affinity-based protein profiling (AfBPP), we identified isocitrate dehydrogenase 2 (IDH2) as a key mitochondrial target of CBN. This finding was further substantiated by siRNA knockdown studies, which revealed that the absence of IDH2 partially phenocopies CBN's effects, validating its role as a critical mediator of CBN's neuroprotective activity.
Collapse
Affiliation(s)
- Eva Schaller
- Julius-Maximilians-Universität Würzburg (JMU), Institute for Pharmacy and Food Chemistry, Pharmaceutical and Medicinal Chemistry, Am Hubland, 97074, Würzburg, Germany
| | - Stephanie Lamer
- Julius-Maximilians-Universität Würzburg (JMU), Rudolf-Virchow-Zentrum - Center for Integrative and Translational Bioimaging, 97080, Würzburg, Germany
| | - Andreas Schlosser
- Julius-Maximilians-Universität Würzburg (JMU), Rudolf-Virchow-Zentrum - Center for Integrative and Translational Bioimaging, 97080, Würzburg, Germany
| | - Christian Stigloher
- Julius-Maximilians-Universität Würzburg (JMU), Biocenter/Theodor-Boveri-Institute, Imaging Core Facility, Am Hubland, 97074, Würzburg, Germany
| | - Pamela Maher
- Cellular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N Torrey Pines Road, La Jolla, California, 92037, USA
| | - Michael Decker
- Julius-Maximilians-Universität Würzburg (JMU), Institute for Pharmacy and Food Chemistry, Pharmaceutical and Medicinal Chemistry, Am Hubland, 97074, Würzburg, Germany
| |
Collapse
|
49
|
Belda H, Bradley D, Christodoulou E, Nofal SD, Broncel M, Jones D, Davies H, Bertran MT, Purkiss AG, Ogrodowicz RW, Joshi D, O'Reilly N, Walport L, Powell A, House D, Kjaer S, Claessens A, Landry CR, Treeck M. The fast-evolving FIKK kinase family of Plasmodium falciparum can be inhibited by a single compound. Nat Microbiol 2025:10.1038/s41564-025-02017-4. [PMID: 40389650 DOI: 10.1038/s41564-025-02017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/14/2025] [Indexed: 05/21/2025]
Abstract
Of 250 Plasmodium species, 6 infect humans, with P. falciparum causing over 95% of 600,000 annual malaria-related deaths. Its pathology arises from host cell remodelling driven by over 400 exported parasite proteins, including the FIKK kinase family. About one million years ago, a bird-infecting Plasmodium species crossed into great apes and a single non-exported FIKK kinase gained an export element. This led to a rapid expansion into 15-21 atypical, exported Ser/Thr effector kinases. Here, using genomic and proteomic analyses, we demonstrate FIKK differentiation via changes in subcellular localization, expression timing and substrate motifs, which supports an individual important role in host-pathogen interactions. Structural data and AlphaFold2 predictions reveal fast-evolving loops in the kinase domain that probably enabled rapid functional diversification for substrate preferences. One FIKK evolved exclusive tyrosine phosphorylation, previously thought absent in Plasmodium. Despite divergence of substrate preferences, the atypical ATP binding pocket is conserved and we identified a single compound that inhibits all FIKKs. A pan-specific inhibitor could reduce resistance development and improve malaria control strategies.
Collapse
Affiliation(s)
- Hugo Belda
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - David Bradley
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Quebec, Canada
- Institut de Biologie Intégrative et des Systems, Université Laval, Québec, Quebec, Canada
- PROTEO, Le Groupement Québécois de Recherche sur la Function, l'Ingénierie et les Applications des Proteins, Université Laval, Québec, Quebec, Canada
| | | | - Stephanie D Nofal
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Malgorzata Broncel
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
| | - David Jones
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
| | - Heledd Davies
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK
| | - M Teresa Bertran
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, UK
| | - Andrew G Purkiss
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Roksana W Ogrodowicz
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Dhira Joshi
- Chemical Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Nicola O'Reilly
- Chemical Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Louise Walport
- Protein-Protein Interaction Laboratory, The Francis Crick Institute, London, UK
| | | | - David House
- CrickGSK Biomedical LinkLabs, GSK, Stevenage, UK
| | - Svend Kjaer
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Antoine Claessens
- LPHI, MIVEGEC, INSERM, CNRS, IRD, University of Montpellier, Montpellier, France
| | - Christian R Landry
- Département de Biochimie, de Microbiologie et de Bio-informatique, Faculté des Sciences et de Génie, Université Laval, Québec, Quebec, Canada
- Institut de Biologie Intégrative et des Systems, Université Laval, Québec, Quebec, Canada
- PROTEO, Le Groupement Québécois de Recherche sur la Function, l'Ingénierie et les Applications des Proteins, Université Laval, Québec, Quebec, Canada
| | - Moritz Treeck
- Signalling in Apicomplexan Parasites Laboratory, The Francis Crick Institute, London, UK.
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal.
| |
Collapse
|
50
|
Tutak K, Broniarek I, Zielezinski A, Niewiadomska D, Skrzypczak T, Baud A, Sobczak K. Insufficiency of 40S ribosomal proteins, RPS26 and RPS25, negatively affects biosynthesis of polyglycine-containing proteins in fragile-X associated conditions. eLife 2025; 13:RP98631. [PMID: 40377206 DOI: 10.7554/elife.98631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025] Open
Abstract
Expansion of CGG repeats (CGGexp) in the 5' untranslated region (5'UTR) of the FMR1 gene underlies the fragile X premutation-associated conditions including tremor/ataxia syndrome, a late-onset neurodegenerative disease and fragile X-associated primary ovarian insufficiency. One common pathomechanism of these conditions is the repeat-associated non-AUG-initiated (RAN) translation of CGG repeats of mutant FMR1 mRNA, resulting in production of FMRpolyG, a toxic protein containing long polyglycine tract. To identify novel modifiers of RAN translation we used an RNA-tagging system and mass spectrometry-based screening. It revealed proteins enriched on CGGexp-containing FMR1 RNA in cellulo, including a ribosomal protein RPS26, a component of the 40 S subunit. We demonstrated that depletion of RPS26 and its chaperone TSR2, modulates FMRpolyG production and its toxicity. We also found that the RPS26 insufficiency impacted translation of limited number of proteins, and 5'UTRs of mRNAs encoding these proteins were short and guanosine and cytosine-rich. Moreover, the silencing of another component of the 40 S subunit, the ribosomal protein RPS25, also induced repression of FMRpolyG biosynthesis. Results of this study suggest that the two 40 S ribosomal proteins and chaperone TSR2 play an important role in noncanonical CGGexp-related RAN translation.
Collapse
Affiliation(s)
- Katarzyna Tutak
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, Poznan, Poland
| | - Izabela Broniarek
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, Poznan, Poland
| | - Andrzej Zielezinski
- Department of Computational Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University Uniwersytetu Poznańskiego 6, Poznan, Poland
| | - Daria Niewiadomska
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, Poznan, Poland
| | - Tomasz Skrzypczak
- Center of Advanced Technology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 10, Poznan, Poland
| | - Anna Baud
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, Poznan, Poland
| | - Krzysztof Sobczak
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Uniwersytetu Poznańskiego 6, Poznan, Poland
| |
Collapse
|