201
|
Proteomics Evidence of a Systemic Response to Desiccation in the Resurrection Plant Haberlea rhodopensis. Int J Mol Sci 2022; 23:ijms23158520. [PMID: 35955654 PMCID: PMC9369045 DOI: 10.3390/ijms23158520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 07/21/2022] [Accepted: 07/26/2022] [Indexed: 02/01/2023] Open
Abstract
Global warming and drought stress are expected to have a negative impact on agricultural productivity. Desiccation-tolerant species, which are able to tolerate the almost complete desiccation of their vegetative tissues, are appropriate models to study extreme drought tolerance and identify novel approaches to improve the resistance of crops to drought stress. In the present study, to better understand what makes resurrection plants extremely tolerant to drought, we performed transmission electron microscopy and integrative large-scale proteomics, including organellar and phosphorylation proteomics, and combined these investigations with previously published transcriptomic and metabolomics data from the resurrection plant Haberlea rhodopensis. The results revealed new evidence about organelle and cell preservation, posttranscriptional and posttranslational regulation, photosynthesis, primary metabolism, autophagy, and cell death in response to desiccation in H. rhodopensis. Different protective intrinsically disordered proteins, such as late embryogenesis abundant (LEA) proteins, thaumatin-like proteins (TLPs), and heat shock proteins (HSPs), were detected. We also found a constitutively abundant dehydrin in H. rhodopensis whose phosphorylation levels increased under stress in the chloroplast fraction. This integrative multi-omics analysis revealed a systemic response to desiccation in H. rhodopensis and certain targets for further genomic and evolutionary studies on DT mechanisms and genetic engineering towards the improvement of drought tolerance in crops.
Collapse
|
202
|
Hernández ÁP, Micaelo A, Piñol R, García-Vaquero ML, Aramayona JJ, Criado JJ, Rodriguez E, Sánchez-Gallego JI, Landeira-Viñuela A, Juanes-Velasco P, Díez P, Góngora R, Jara-Acevedo R, Orfao A, Miana-Mena J, Muñoz MJ, Villanueva S, Millán Á, Fuentes M. Comprehensive and systematic characterization of multi-functionalized cisplatin nano-conjugate: from the chemistry and proteomic biocompatibility to the animal model. J Nanobiotechnology 2022; 20:341. [PMID: 35858906 PMCID: PMC9301860 DOI: 10.1186/s12951-022-01546-y] [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: 04/26/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Background Nowadays, nanoparticles (NPs) have evolved as multifunctional systems combining different custom anchorages which opens a wide range of applications in biomedical research. Thus, their pharmacological involvements require more comprehensive analysis and novel nanodrugs should be characterized by both chemically and biological point of view. Within the wide variety of biocompatible nanosystems, iron oxide nanoparticles (IONPs) present mostly of the required features which make them suitable for multifunctional NPs with many biopharmaceutical applications. Results Cisplatin-IONPs and different functionalization stages have been broadly evaluated. The potential application of these nanodrugs in onco-therapies has been assessed by studying in vitro biocompatibility (interactions with environment) by proteomics characterization the determination of protein corona in different proximal fluids (human plasma, rabbit plasma and fetal bovine serum),. Moreover, protein labeling and LC–MS/MS analysis provided more than 4000 proteins de novo synthetized as consequence of the nanodrugs presence defending cell signaling in different tumor cell types (data available via ProteomeXchanges with identified PXD026615). Further in vivo studies have provided a more integrative view of the biopharmaceutical perspectives of IONPs. Conclusions Pharmacological proteomic profile different behavior between species and different affinity of protein coating layers (soft and hard corona). Also, intracellular signaling exposed differences between tumor cell lines studied. First approaches in animal model reveal the potential of theses NPs as drug delivery vehicles and confirm cisplatin compounds as strengthened antitumoral agents.
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01546-y.
Collapse
Affiliation(s)
- Ángela-Patricia Hernández
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,Department of Pharmaceutical Sciences. Organic Chemistry Section. Faculty of Pharmacy, University of Salamanca, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Ania Micaelo
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Rafael Piñol
- INMA, Institute of Nanoscience and Materials of Aragon, CSIC-University of Zaragoza, 50018, Saragossa, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Marina L García-Vaquero
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - José J Aramayona
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Julio J Criado
- Department of Inorganic Chemistry, Faculty of Chemical Sciences, Plaza de los Caídos S/N, 37008, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Emilio Rodriguez
- Department of Inorganic Chemistry, Faculty of Chemical Sciences, Plaza de los Caídos S/N, 37008, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - José Ignacio Sánchez-Gallego
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Alicia Landeira-Viñuela
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Pablo Juanes-Velasco
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Paula Díez
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Rafael Góngora
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Ricardo Jara-Acevedo
- ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Javier Miana-Mena
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - María Jesús Muñoz
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Sergio Villanueva
- Department of Pharmacology and Physiology, University of Zaragoza, Zaragoza, Spain.,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Ángel Millán
- INMA, Institute of Nanoscience and Materials of Aragon, CSIC-University of Zaragoza, 50018, Saragossa, Spain. .,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain.
| | - Manuel Fuentes
- Department of Medicine and General Cytometry Service-Nucleus, CIBERONC CB16/12/00400, Cancer Research Centre, (IBMCC/CSIC/USAL/IBSAL), University of Salamanca-CSIC, IBSAL, Campus Miguel de Unamuno s/n, 37007, Salamanca, Spain. .,ImmunoStep, SL, Edificio Centro de Investigación del Cáncer, University of Salamanca, Avda. Coimbra s/n, Campus Miguel de Unamuno, 37007, Salamanca, Spain. .,Proteomics Unit, Cancer Research Centre (IBMCC/CSIC/USAL/IBSAL), 37007, Salamanca, Spain.
| |
Collapse
|
203
|
Mestre-Farràs N, Guerrero S, Bley N, Rivero E, Coll O, Borràs E, Sabidó E, Indacochea A, Casillas-Serra C, Järvelin AI, Oliva B, Castello A, Hüttelmaier S, Gebauer F. Melanoma RBPome identification reveals PDIA6 as an unconventional RNA-binding protein involved in metastasis. Nucleic Acids Res 2022; 50:8207-8225. [PMID: 35848924 PMCID: PMC9371929 DOI: 10.1093/nar/gkac605] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 06/10/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
RNA-binding proteins (RBPs) have been relatively overlooked in cancer research despite their contribution to virtually every cancer hallmark. Here, we use RNA interactome capture (RIC) to characterize the melanoma RBPome and uncover novel RBPs involved in melanoma progression. Comparison of RIC profiles of a non-tumoral versus a metastatic cell line revealed prevalent changes in RNA-binding capacities that were not associated with changes in RBP levels. Extensive functional validation of a selected group of 24 RBPs using five different in vitro assays unveiled unanticipated roles of RBPs in melanoma malignancy. As proof-of-principle we focused on PDIA6, an ER-lumen chaperone that displayed a novel RNA-binding activity. We show that PDIA6 is involved in metastatic progression, map its RNA-binding domain, and find that RNA binding is required for PDIA6 tumorigenic properties. These results exemplify how RIC technologies can be harnessed to uncover novel vulnerabilities of cancer cells.
Collapse
Affiliation(s)
- Neus Mestre-Farràs
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Santiago Guerrero
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Nadine Bley
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Ezequiel Rivero
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Olga Coll
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Eva Borràs
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain.,Department of Health and Experimental Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Eduard Sabidó
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain.,Department of Health and Experimental Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Alberto Indacochea
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Carlos Casillas-Serra
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain
| | - Aino I Järvelin
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Baldomero Oliva
- Department of Health and Experimental Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Alfredo Castello
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Section for Molecular Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany
| | - Fátima Gebauer
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, 08003 Barcelona, Spain.,Department of Health and Experimental Sciences, Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| |
Collapse
|
204
|
Smertina E, Carroll AJ, Boileau J, Emmott E, Jenckel M, Vohra H, Rolland V, Hands P, Hayashi J, Neave MJ, Liu JW, Hall RN, Strive T, Frese M. Lagovirus Non-structural Protein p23: A Putative Viroporin That Interacts With Heat Shock Proteins and Uses a Disulfide Bond for Dimerization. Front Microbiol 2022; 13:923256. [PMID: 35923397 PMCID: PMC9340658 DOI: 10.3389/fmicb.2022.923256] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/16/2022] [Indexed: 12/03/2022] Open
Abstract
The exact function(s) of the lagovirus non-structural protein p23 is unknown as robust cell culture systems for the Rabbit haemorrhagic disease virus (RHDV) and other lagoviruses have not been established. Instead, a range of in vitro and in silico models have been used to study p23, revealing that p23 oligomerizes, accumulates in the cytoplasm, and possesses a conserved C-terminal region with two amphipathic helices. Furthermore, the positional homologs of p23 in other caliciviruses have been shown to possess viroporin activity. Here, we report on the mechanistic details of p23 oligomerization. Site-directed mutagenesis revealed the importance of an N-terminal cysteine for dimerization. Furthermore, we identified cellular interactors of p23 using stable isotope labeling with amino acids in cell culture (SILAC)-based proteomics; heat shock proteins Hsp70 and 110 interact with p23 in transfected cells, suggesting that they ‘chaperone’ p23 proteins before their integration into cellular membranes. We investigated changes to the global transcriptome and proteome that occurred in infected rabbit liver tissue and observed changes to the misfolded protein response, calcium signaling, and the regulation of the endoplasmic reticulum (ER) network. Finally, flow cytometry studies indicate slightly elevated calcium concentrations in the cytoplasm of p23-transfected cells. Taken together, accumulating evidence suggests that p23 is a viroporin that might form calcium-conducting channels in the ER membranes.
Collapse
Affiliation(s)
- Elena Smertina
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
- Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
- Centre for Invasive Species Solutions, Canberra, ACT, Australia
| | - Adam J. Carroll
- RSB/RSC Joint Mass Spectrometry Facility, Research School of Chemistry, Australian National University, Acton, ACT, Australia
| | - Joseph Boileau
- RSB/RSC Joint Mass Spectrometry Facility, Research School of Chemistry, Australian National University, Acton, ACT, Australia
| | - Edward Emmott
- Centre for Proteome Research, Department of Biochemistry & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Maria Jenckel
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
| | - Harpreet Vohra
- Imaging and Cytometry Facility, John Curtin School of Medical Research, Acton, ACT, Australia
| | - Vivien Rolland
- Agriculture and Food, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
| | - Philip Hands
- Agriculture and Food, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
| | - Junna Hayashi
- Research School of Chemistry, Australian National University, Acton, ACT, Australia
| | - Matthew J. Neave
- Australian Centre for Disease Preparedness, Commonwealth Scientific and Industrial Research Organisation, Geelong, VIC, Australia
| | - Jian-Wei Liu
- Land and Water, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
| | - Robyn N. Hall
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
- Centre for Invasive Species Solutions, Canberra, ACT, Australia
| | - Tanja Strive
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
- Centre for Invasive Species Solutions, Canberra, ACT, Australia
| | - Michael Frese
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
- Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
- *Correspondence: Michael Frese,
| |
Collapse
|
205
|
Ishida M, Maki Y, Ninomiya A, Takada Y, Campeau P, Kinoshita T, Murakami Y. Ethanolamine-phosphate on the second mannose is a preferential bridge for some GPI-anchored proteins. EMBO Rep 2022; 23:e54352. [PMID: 35603428 PMCID: PMC9253782 DOI: 10.15252/embr.202154352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/13/2022] [Accepted: 04/25/2022] [Indexed: 09/10/2023] Open
Abstract
Glycosylphosphatidylinositols (GPIs) are glycolipids that anchor many proteins (GPI-APs) on the cell surface. The core glycan of GPI precursor has three mannoses, which in mammals, are all modified by ethanolamine-phosphate (EthN-P). It is postulated that EthN-P on the third mannose (EthN-P-Man3) is the bridge between GPI and the protein and the second (EthN-P-Man2) is removed after GPI-protein attachment. However, EthN-P-Man2 may not be always transient, as mutations of PIGG, the enzyme that transfers EthN-P to Man2, result in inherited GPI deficiencies (IGDs), characterized by neuronal dysfunctions. Here, we show that EthN-P on Man2 is the preferential bridge in some GPI-APs, among them, the Ect-5'-nucleotidase and Netrin G2. We find that CD59, a GPI-AP, is attached via EthN-P-Man2 both in PIGB-knockout cells, in which GPI lacks Man3, and with a small fraction in wild-type cells. Our findings modify the current view of GPI anchoring and provide a mechanistic basis for IGDs caused by PIGG mutations.
Collapse
Affiliation(s)
- Mizuki Ishida
- Yabumoto Department of Intractable Disease ResearchResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
| | - Yuta Maki
- Department of ChemistryOsaka UniversityToyonakaJapan
- Project Research Center for Fundamental SciencesGraduate School of ScienceOsaka UniversityToyonakaJapan
| | - Akinori Ninomiya
- Central Instrumentation LaboratoryResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
| | - Yoko Takada
- WPI Immunology Frontier Research CenterOsaka UniversitySuitaJapan
| | - Philippe Campeau
- Department of PediatricsCHU Sainte‐Justine and University of MontrealMontrealQCCanada
| | - Taroh Kinoshita
- Yabumoto Department of Intractable Disease ResearchResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
- WPI Immunology Frontier Research CenterOsaka UniversitySuitaJapan
- Center for Infectious Disease Education and ResearchOsaka UniversitySuitaJapan
| | - Yoshiko Murakami
- Yabumoto Department of Intractable Disease ResearchResearch Institute for Microbial DiseasesOsaka UniversitySuitaJapan
- WPI Immunology Frontier Research CenterOsaka UniversitySuitaJapan
| |
Collapse
|
206
|
Bauzá-Martinez J, Armony G, Pronker MF, Wu W. Characterization of protein complexes in extracellular vesicles by intact extracellular vesicle crosslinking mass spectrometry (iEVXL). J Extracell Vesicles 2022; 11:e12245. [PMID: 35918900 PMCID: PMC9346492 DOI: 10.1002/jev2.12245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/05/2022] [Accepted: 06/25/2022] [Indexed: 11/15/2022] Open
Abstract
Extracellular vesicles (EVs) are blood‐borne messengers that coordinate signalling between different tissues and organs in the body. The specificity of such crosstalk is determined by preferential EV docking to target sites, as mediated through protein‐protein interactions. As such, the need to structurally characterize the EV surface precedes further understanding of docking selectivity and recipient‐cell uptake mechanisms. Here, we describe an intact extracellular vesicle crosslinking mass spectrometry (iEVXL) method that can be applied for structural characterization of protein complexes in EVs. By using a partially membrane‐permeable disuccinimidyl suberate crosslinker, proteins on the EV outer‐surface and inside EVs can be immobilized together with their interacting partners. This not only provides covalent stabilization of protein complexes before extraction from the membrane‐enclosed environment, but also generates a set of crosslinking distance restraints that can be used for structural modelling and comparative screening of changes in EV protein assemblies. Here we demonstrate iEVXL as a powerful approach to reveal high‐resolution information, about protein determinants that govern EV docking and signalling, and as a crucial aid in modelling docking interactions.
Collapse
Affiliation(s)
- Julia Bauzá-Martinez
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Gad Armony
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Matti F Pronker
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Wei Wu
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands.,Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Department of Pharmacy, National University of Singapore, Singapore, Singapore
| |
Collapse
|
207
|
Verreault M, Segoviano Vilchis I, Rosenberg S, Lemaire N, Schmitt C, Guehennec J, Royer-Perron L, Thomas JL, Lam TT, Dingli F, Loew D, Ducray F, Paris S, Carpentier C, Marie Y, Laigle-Donadey F, Rousseau A, Pigat N, Boutillon F, Bielle F, Mokhtari K, Frank SJ, de Reyniès A, Hoang-Xuan K, Sanson M, Goffin V, Idbaih A. Identification of growth hormone receptor as a relevant target for precision medicine in low-EGFR expressing glioblastoma. Clin Transl Med 2022; 12:e939. [PMID: 35808822 PMCID: PMC9270581 DOI: 10.1002/ctm2.939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 05/30/2022] [Accepted: 06/05/2022] [Indexed: 11/11/2022] Open
Abstract
Objective New therapeutic approaches are needed to improve the prognosis of glioblastoma (GBM) patients. Methods With the objective of identifying alternative oncogenic mechanisms to abnormally activated epidermal growth factor receptor (EGFR) signalling, one of the most common oncogenic mechanisms in GBM, we performed a comparative analysis of gene expression profiles in a series of 54 human GBM samples. We then conducted gain of function as well as genetic and pharmocological inhibition assays in GBM patient‐derived cell lines to functionnally validate our finding. Results We identified that growth hormone receptor (GHR) signalling defines a distinct molecular subset of GBMs devoid of EGFR overexpression. GHR overexpression was detected in one third of patients and was associated with low levels of suppressor of cytokine signalling 2 (SOCS2) expression due to SOCS2 promoter hypermethylation. In GBM patient‐derived cell lines, GHR signalling modulates the expression of proteins involved in cellular movement, promotes cell migration, invasion and proliferation in vitro and promotes tumourigenesis, tumour growth, and tumour invasion in vivo. GHR genetic and pharmacological inhibition reduced cell proliferation and migration in vitro. Conclusion This study pioneers a new field of investigation to improve the prognosis of GBM patients.
Collapse
Affiliation(s)
- Maïté Verreault
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Irma Segoviano Vilchis
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Shai Rosenberg
- Laboratory for Cancer Computational Biology & Gaffin Center for Neuro-Oncology, Hadassah - Hebrew University Medical Center, Jerusalem, Israel
| | - Nolwenn Lemaire
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Charlotte Schmitt
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Jérémy Guehennec
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Louis Royer-Perron
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Jean-Léon Thomas
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France.,Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - TuKiet T Lam
- Mass Spectrometry & Proteomics Resource, Keck Biotechnology Resource Laboratory, New Haven, Connecticut, USA.,Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Florent Dingli
- Institut Curie, Centre de Recherche, PSL Research University, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | - Damarys Loew
- Institut Curie, Centre de Recherche, PSL Research University, Laboratoire de Spectrométrie de Masse Protéomique, Paris, France
| | | | - Sophie Paris
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Catherine Carpentier
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Yannick Marie
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Florence Laigle-Donadey
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Audrey Rousseau
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France.,DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Natascha Pigat
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Paris, France
| | - Florence Boutillon
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Paris, France
| | - Franck Bielle
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Karima Mokhtari
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stuart J Frank
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama, Birmingham, Alabama, USA.,Endocrinology Section, Medical Service, Birmingham VA Medical Center, Birmingham, Alabama, USA
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs (CIT), Ligue Nationale Contre le Cancer, Service de Bioinformatique, Paris, France
| | - Khê Hoang-Xuan
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Marc Sanson
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Vincent Goffin
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Paris, France
| | - Ahmed Idbaih
- DMU Neurosciences, Service de Neurologie 2-Mazarin, Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| |
Collapse
|
208
|
Hollenstein DM, Veis J, Romanov N, Gérecová G, Ogris E, Hartl M, Ammerer G, Reiter W. PP2A Rts1 antagonizes Rck2-mediated hyperosmotic stress signaling in yeast. Microbiol Res 2022; 260:127031. [PMID: 35461031 DOI: 10.1016/j.micres.2022.127031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 11/21/2022]
Abstract
In Saccharomyces cerevisiae, impairment of protein phosphatase PP2ARts1 leads to temperature and hyperosmotic stress sensitivity, yet the underlying mechanism and the scope of action of the phosphatase in the stress response remain elusive. Using a quantitative mass spectrometry-based approach we have identified a set of putative substrate proteins that show both hyperosmotic stress- and PP2ARts1-dependent changes in their phosphorylation pattern. A comparative analysis with published MS-shotgun data revealed that the phosphorylation status of many of these sites is regulated by the MAPKAP kinase Rck2, suggesting that the phosphatase antagonizes Rck2 signaling. Detailed gel mobility shift assays and protein-protein interaction analysis strongly indicate that Rck2 activity is directly regulated by PP2ARts1 via a SLiM B56-family interaction motif, revealing how PP2ARts1 influences the response to hyperosmotic stress in Yeast.
Collapse
Affiliation(s)
- D M Hollenstein
- Department of Biochemistry and Cell Biology, Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - J Veis
- Department of Biochemistry and Cell Biology, Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria; Center for Medical Biochemistry, Max Perutz Labs, Medical University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - N Romanov
- Max Planck Institute of Biophysics, Max-von-Laue Straße 3, 60438 Frankfurt am Main, Germany
| | - G Gérecová
- Department of Biochemistry and Cell Biology, Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - E Ogris
- Center for Medical Biochemistry, Max Perutz Labs, Medical University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - M Hartl
- Mass Spectrometry Facility, Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - G Ammerer
- Department of Biochemistry and Cell Biology, Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria
| | - W Reiter
- Department of Biochemistry and Cell Biology, Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria; Mass Spectrometry Facility, Max Perutz Labs, University of Vienna, Vienna BioCenter, Dr. Bohr-Gasse 9, 1030 Vienna, Austria.
| |
Collapse
|
209
|
Kumar A, Nonnis S, Castellano I, AbdElgawad H, Beemster GTS, Buia MC, Maffioli E, Tedeschi G, Palumbo A. Molecular response of Sargassum vulgare to acidification at volcanic CO 2 vents: Insights from proteomic and metabolite analyses. Mol Ecol 2022; 31:3844-3858. [PMID: 35635253 DOI: 10.1111/mec.16553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 11/30/2022]
Abstract
Ocean acidification is impacting marine life all over the world. Understanding how species can cope with the changes in seawater carbonate chemistry represents a challenging issue. We addressed this topic using underwater CO2 vents that naturally acidify some marine areas off the island of Ischia. In the most acidified area of the vents, having a mean pH value of 6.7, comparable to far-future predicted acidification scenarios (by 2300), the biomass is dominated by the brown alga Sargassum vulgare. The novelty of the present study is the characterization of the S. vulgare proteome together with metabolite analyses to identify the key proteins, metabolites, and pathways affected by ocean acidification. A total of 367 and 387 proteins were identified in populations grown at pH that approximates the current global average (8.1) and acidified sites, respectively. Analysis of their relative abundance revealed that 304 proteins are present in samples from both sites: 111 proteins are either higher or exclusively present under acidified conditions, whereas 120 proteins are either lower or present only under control conditions. Functionally, under acidification, a decrease in proteins related to translation and post-translational processes and an increase of proteins involved in photosynthesis, glycolysis, oxidation-reduction processes, and protein folding were observed. In addition, small-molecule metabolism was affected, leading to a decrease of some fatty acids and antioxidant compounds under acidification. Overall, the results obtained by proteins and metabolites analyses, integrated with previous transcriptomic, physiological, and biochemical studies, allowed us to delineate the molecular strategies adopted by S. vulgare to grow in future acidified environments, including an increase of proteins involved in energetic metabolism, oxidation-reduction processes, and protein folding at the expense of proteins involved in translation and post-translational processes.
Collapse
Affiliation(s)
- Amit Kumar
- Stazione Zoologica Anton Dohrn, Department of Integrative Marine Ecology, Marine Research Center, Naples, Italy
- Centre for Climate Change Studies, Sathyabama Institute of Science and Technology, Chennai, India
| | - Simona Nonnis
- Department of Veterinary Medicine and Animal Science (DIVAS), Università degli Studi di Milano, Milan, Italy
- CRC "Innovation for well-being and environment" (I-WE), Università degli Studi di Milano, Milan, Italy
| | - Immacolata Castellano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, Naples, Italy
- Stazione Zoologica Anton Dohrn, Department of Biology and Evolution of Marine Organisms, Naples, Italy
| | - Hamada AbdElgawad
- Department of Botany, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
- Integrated Molecular Plant Physiology Research Group (IMPRES), Department of Biology, University of Antwerp, Antwerp, Belgium
| | - Gerrit T S Beemster
- Integrated Molecular Plant Physiology Research Group (IMPRES), Department of Biology, University of Antwerp, Antwerp, Belgium
| | - Maria Cristina Buia
- Stazione Zoologica Anton Dohrn, Department of Integrative Marine Ecology, Marine Research Center, Naples, Italy
| | - Elisa Maffioli
- Department of Veterinary Medicine and Animal Science (DIVAS), Università degli Studi di Milano, Milan, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science (DIVAS), Università degli Studi di Milano, Milan, Italy
- CRC "Innovation for well-being and environment" (I-WE), Università degli Studi di Milano, Milan, Italy
| | - Anna Palumbo
- Stazione Zoologica Anton Dohrn, Department of Biology and Evolution of Marine Organisms, Naples, Italy
| |
Collapse
|
210
|
Domenzain I, Sánchez B, Anton M, Kerkhoven EJ, Millán-Oropeza A, Henry C, Siewers V, Morrissey JP, Sonnenschein N, Nielsen J. Reconstruction of a catalogue of genome-scale metabolic models with enzymatic constraints using GECKO 2.0. Nat Commun 2022; 13:3766. [PMID: 35773252 PMCID: PMC9246944 DOI: 10.1038/s41467-022-31421-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/16/2022] [Indexed: 01/08/2023] Open
Abstract
Genome-scale metabolic models (GEMs) have been widely used for quantitative exploration of the relation between genotype and phenotype. Streamlined integration of enzyme constraints and proteomics data into such models was first enabled by the GECKO toolbox, allowing the study of phenotypes constrained by protein limitations. Here, we upgrade the toolbox in order to enhance models with enzyme and proteomics constraints for any organism with a compatible GEM reconstruction. With this, enzyme-constrained models for the budding yeasts Saccharomyces cerevisiae, Yarrowia lipolytica and Kluyveromyces marxianus are generated to study their long-term adaptation to several stress factors by incorporation of proteomics data. Predictions reveal that upregulation and high saturation of enzymes in amino acid metabolism are common across organisms and conditions, suggesting the relevance of metabolic robustness in contrast to optimal protein utilization as a cellular objective for microbial growth under stress and nutrient-limited conditions. The functionality of GECKO is expanded with an automated framework for continuous and version-controlled update of enzyme-constrained GEMs, also producing such models for Escherichia coli and Homo sapiens. In this work, we facilitate the utilization of enzyme-constrained GEMs in basic science, metabolic engineering and synthetic biology purposes.
Collapse
Affiliation(s)
- Iván Domenzain
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Benjamín Sánchez
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Mihail Anton
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
- Department of Biology and Biological Engineering, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Chalmers University of Technology, Kemivägen 10, SE-412 58, Gothenburg, Sweden
| | - Eduard J Kerkhoven
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Aarón Millán-Oropeza
- Plateforme d'analyse protéomique Paris Sud-Ouest (PAPPSO), INRAE, MICALIS Institute, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Céline Henry
- Plateforme d'analyse protéomique Paris Sud-Ouest (PAPPSO), INRAE, MICALIS Institute, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Verena Siewers
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - John P Morrissey
- School of Microbiology, Environmental Research Institute and APC Microbiome Ireland, University College Cork, T12 K8AF, Cork, Ireland
| | - Nikolaus Sonnenschein
- Department of Biotechnology and Biomedicine, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden.
- Novo Nordisk Foundation Center for Biosustainability, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden.
- BioInnovation Institute, Ole Maaløes Vej 3, 2200, Copenhagen, Denmark.
| |
Collapse
|
211
|
Manganese Modulates Metabolic Activity and Redox Homeostasis in Translationally Blocked Lactococcus cremoris, Impacting Metabolic Persistence, Cell Culturability, and Flavor Formation. Microbiol Spectr 2022; 10:e0270821. [PMID: 35638825 PMCID: PMC9241929 DOI: 10.1128/spectrum.02708-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Manganese (Mn) is an essential trace element that is supplemented in microbial media with varying benefits across species and growth conditions. We found that growth of Lactococcus cremoris was unaffected by manganese omission from the growth medium. The main proteome adaptation to manganese omission involved increased manganese transporter production (up to 2,000-fold), while the remaining 10 significant proteome changes were between 1.4- and 4-fold. Further investigation in translationally blocked (TB), nongrowing cells showed that Mn supplementation (20 μM) led to approximately 1.5 X faster acidification compared with Mn-free conditions. However, this faster acidification stagnated within 24 h, likely due to draining of intracellular NADH that coincides with substantial loss of culturability. Conversely, without manganese, nongrowing cells persisted to acidify for weeks, albeit at a reduced rate, but maintaining redox balance and culturability. Strikingly, despite being unculturable, α-keto acid-derived aldehydes continued to accumulate in cells incubated in the presence of manganese, whereas without manganese cells predominantly formed the corresponding alcohols. This is most likely reflecting NADH availability for the alcohol dehydrogenase-catalyzed conversion. Overall, manganese influences the lactococcal acidification rate, and flavor formation capacity in a redox dependent manner. These are important industrial traits especially during cheese ripening, where cells are in a non-growing, often unculturable state. IMPORTANCE In nature as well as in various biotechnology applications, microorganisms are often in a nongrowing state and their metabolic persistence determines cell survival and functionality. Industrial examples are dairy fermentations where bacteria remain active during the ripening phases that can take up to months and even years. Here we investigated environmental factors that can influence lactococcal metabolic persistence throughout such prolonged periods. We found that in the absence of manganese, acidification of nongrowing cells remained active for weeks while in the presence of manganese it stopped within 1 day. The latter coincided with the accumulation of amino acid derived volatile metabolites. Based on metabolic conversions, proteome analysis, and a reporter assay, we demonstrated that the manganese elicited effects were NADH dependent. Overall the results show the effect of environmental modulation on prolonged cell-based catalysis, which is highly relevant to non-growing cells in nature and biotechnological applications.
Collapse
|
212
|
Crizotinib Shows Antibacterial Activity against Gram-Positive Bacteria by Reducing ATP Production and Targeting the CTP Synthase PyrG. Microbiol Spectr 2022; 10:e0088422. [PMID: 35674439 PMCID: PMC9241945 DOI: 10.1128/spectrum.00884-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infections caused by drug-resistant bacteria are a serious threat to public health worldwide, and the discovery of novel antibacterial compounds is urgently needed. Here, we screened an FDA-approved small-molecule library and found that crizotinib possesses good antimicrobial efficacy against Gram-positive bacteria. Crizotinib was found to increase the survival rate of mice infected with bacteria and decrease pulmonary inflammation activity in an animal model. Furthermore, it showed synergy with clindamycin and gentamicin. Importantly, the Gram-positive bacteria showed a low tendency to develop resistance to crizotinib. Mechanistically, quantitative proteomics and biochemical validation experiments indicated that crizotinib exerted its antibacterial effects by reducing ATP production and pyrimidine metabolism. A drug affinity responsive target stability study suggested crizotinib targets the CTP synthase PyrG, which subsequently disturbs pyrimidine metabolism and eventually reduces DNA synthesis. Subsequent molecular dynamics analysis showed that crizotinib binding occurs in close proximity to the ATP binding pocket of PyrG and causes loss of function of this CTP synthase. Crizotinib is a promising antimicrobial agent and provides a novel choice for the development of treatment for Gram-positive infections. IMPORTANCE Infections caused by drug-resistant bacteria are a serious problem worldwide. Therefore, there is an urgent need to find novel drugs with good antibacterial activity against multidrug-resistant bacteria. In this study, we found that a repurposed drug, crizotinib, exhibits excellent antibacterial activity against drug-resistant bacteria both in vivo and in vitro via suppressing ATP production and pyrimidine metabolism. Crizotinib was found to disturb pyrimidine metabolism by targeting the CTP synthase PyrG, thus reducing DNA synthesis. This unique mechanism of action may explain the decreased development of resistance by Staphylococcus aureus to crizotinib. This study provides a potential option for the treatment of drug-resistant bacterial infections in the future.
Collapse
|
213
|
The Phenylacetic Acid Catabolic Pathway Regulates Antibiotic and Oxidative Stress Responses in Acinetobacter. mBio 2022; 13:e0186321. [PMID: 35467424 PMCID: PMC9239106 DOI: 10.1128/mbio.01863-21] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The opportunistic pathogen Acinetobacter baumannii is responsible for a wide range of infections that are becoming increasingly difficult to treat due to extremely high rates of multidrug resistance. Acinetobacter's pathogenic potential is thought to rely on a "persist and resist" strategy that facilitates its remarkable ability to survive under a variety of harsh conditions. The paa operon is involved in the catabolism of phenylacetic acid (PAA), an intermediate in phenylalanine degradation, and is the most differentially regulated pathway under many environmental conditions. We found that, under subinhibitory concentrations of antibiotics, A. baumannii upregulates expression of the paa operon while simultaneously repressing chaperone-usher Csu pilus expression and biofilm formation. These phenotypes are reverted either by exogenous addition of PAA and its nonmetabolizable derivative 4-fluoro-PAA or by a mutation that blocks PAA degradation. Interference with PAA degradation increases susceptibility to antibiotics and hydrogen peroxide treatment. Transcriptomic and proteomic analyses identified a subset of genes and proteins whose expression is affected by addition of PAA or disruption of the paa pathway. Finally, we demonstrated that blocking PAA catabolism results in attenuated virulence in a murine catheter-associated urinary tract infection (CAUTI) model. We conclude that the paa operon is part of a regulatory network that responds to antibiotic and oxidative stress and is important for virulence. PAA has known regulatory functions in plants, and our experiments suggest that PAA is a cross-kingdom signaling molecule. Interference with this pathway may lead, in the future, to novel therapeutic strategies against A. baumannii infections. IMPORTANCE Acinetobacter baumannii causes a wide range of infections that are difficult to treat due to increasing rates of multidrug resistance; however, the mechanisms that this pathogen uses to respond to stress are poorly understood. Here, we describe a new mechanism of stress signaling in Acinetobacter that is mediated by the metabolite phenylacetic acid (PAA). We found that disrupting PAA catabolism interfered with A. baumannii's ability to adapt to stress, leading to decreased antibiotic tolerance and hydrogen peroxide resistance. We propose that investigating this stress response could lead to the development of novel therapeutics. In fact, PAA derivatives constitute a group of FDA-approved nonsteroidal anti-inflammatory drugs that could potentially be repurposed as antivirulence therapies to target multidrug-resistant Acinetobacter infections.
Collapse
|
214
|
InvL, an Invasin-Like Adhesin, Is a Type II Secretion System Substrate Required for Acinetobacter baumannii Uropathogenesis. mBio 2022; 13:e0025822. [PMID: 35638734 PMCID: PMC9245377 DOI: 10.1128/mbio.00258-22] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Acinetobacter baumannii is an opportunistic pathogen of growing concern, as isolates are commonly multidrug resistant. While A. baumannii is most frequently associated with pulmonary infections, a significant proportion of clinical isolates come from urinary sources, highlighting its uropathogenic potential. The type II secretion system (T2SS) of commonly used model Acinetobacter strains is important for virulence in various animal models, but the potential role of the T2SS in urinary tract infection (UTI) remains unknown. Here, we used a catheter-associated UTI (CAUTI) model to demonstrate that a modern urinary isolate, UPAB1, requires the T2SS for full virulence. A proteomic screen to identify putative UPAB1 T2SS effectors revealed an uncharacterized lipoprotein with structural similarity to the intimin-invasin family, which serve as type V secretion system (T5SS) adhesins required for the pathogenesis of several bacteria. This protein, designated InvL, lacked the β-barrel domain associated with T5SSs but was confirmed to require the T2SS for both surface localization and secretion. This makes InvL the first identified T2SS effector belonging to the intimin-invasin family. InvL was confirmed to be an adhesin, as the protein bound to extracellular matrix components and mediated adhesion to urinary tract cell lines in vitro. Additionally, the invL mutant was attenuated in the CAUTI model, indicating a role in Acinetobacter uropathogenesis. Finally, bioinformatic analyses revealed that InvL is present in nearly all clinical isolates belonging to international clone 2, a lineage of significant clinical importance. In all, we conclude that the T2SS substrate InvL is an adhesin required for A. baumannii uropathogenesis. IMPORTANCE While pathogenic Acinetobacter can cause various infections, we recently found that 20% of clinical isolates come from urinary sources. Despite the clinical relevance of Acinetobacter as a uropathogen, few virulence factors involved in urinary tract colonization have been defined. Here, we identify a novel type II secretion system effector, InvL, which is required for full uropathogenesis by a modern urinary isolate. Although InvL has predicted structural similarity to the intimin-invasin family of autotransporter adhesins, InvL is predicted to be anchored to the membrane as a lipoprotein. Similar to other invasin homologs, however, we demonstrate that InvL is a bona fide adhesin capable of binding extracellular matrix components and mediating adhesion to urinary tract cell lines. In all, this work establishes InvL as an adhesin important for Acinetobacter's urinary tract virulence and represents the first report of a type II secretion system effector belonging to the intimin-invasin family.
Collapse
|
215
|
Griesser E, Schönberger T, Stierstorfer B, Wyatt H, Rist W, Lamla T, Thomas MJ, Lamb D, Geillinger-Kästle KE. Characterization of a flexible AAV-DTR/DT mouse model of acute epithelial lung injury. Am J Physiol Lung Cell Mol Physiol 2022; 323:L206-L218. [PMID: 35762632 DOI: 10.1152/ajplung.00364.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Animal models are important to mimic certain pathways or biological aspects of human pathologies including acute and chronic pulmonary diseases. We developed a novel and flexible mouse model of acute epithelial lung injury based on adeno-associated virus (AAV) variant 6.2 mediated expression of the human diphtheria toxin receptor (DTR). Following intratracheal administration of diphtheria toxin (DT), a cell-specific death of bronchial and alveolar epithelial cells can be observed. In contrast to other lung injury models, the here described mouse model provides the possibility of targeted injury using specific tropisms of AAV vectors or cell type specific promotors to drive the human DTR expression. Also, generation of cell specific mouse lines is not required. Detailed characterization of the AAV-DTR/DT mouse model including titration of viral genome (vg) load and administered DT amount revealed increasing cell numbers in bronchoalveolar lavage (BAL; macrophages, neutrophils, and unspecified cells) and elevation of degenerated cells and infiltrated leukocytes in lung tissue, dependent of vg load and DT dose. Cytokine levels in BAL fluid showed different patterns with higher vg load, e.g. IFNγ, TNFα, and IP10 increasing and IL-5 and IL-6 decreasing, while lung function was not affected. Additionally, laser-capture microdissection (LCM)-based proteomics of bronchial epithelium and alveolar tissue revealed upregulated immune and inflammatory response in all regions and extracellular matrix deposition in infiltrated alveoli. Overall, our novel AAV-DTR/DT model allows investigation of repair mechanisms following epithelial injury and resembles specific mechanistic aspects of acute and chronic pulmonary diseases.
Collapse
Affiliation(s)
- Eva Griesser
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Tanja Schönberger
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Birgit Stierstorfer
- Non-clinical Drug Safety, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Hannah Wyatt
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Wolfgang Rist
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Thorsten Lamla
- Drug Discovery Sciences, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany, Germany
| | - Matthew James Thomas
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany.,University of Bath, Bath, United Kingdom
| | - David Lamb
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany
| | - Kerstin E Geillinger-Kästle
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH and Co. KG, Biberach an der Riss, Germany
| |
Collapse
|
216
|
Depierreux DM, Altenburg AF, Soday L, Fletcher-Etherington A, Antrobus R, Ferguson BJ, Weekes MP, Smith GL. Selective modulation of cell surface proteins during vaccinia infection: A resource for identifying viral immune evasion strategies. PLoS Pathog 2022; 18:e1010612. [PMID: 35727847 PMCID: PMC9307158 DOI: 10.1371/journal.ppat.1010612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 07/22/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022] Open
Abstract
The interaction between immune cells and virus-infected targets involves multiple plasma membrane (PM) proteins. A systematic study of PM protein modulation by vaccinia virus (VACV), the paradigm of host regulation, has the potential to reveal not only novel viral immune evasion mechanisms, but also novel factors critical in host immunity. Here, >1000 PM proteins were quantified throughout VACV infection, revealing selective downregulation of known T and NK cell ligands including HLA-C, downregulation of cytokine receptors including IFNAR2, IL-6ST and IL-10RB, and rapid inhibition of expression of certain protocadherins and ephrins, candidate activating immune ligands. Downregulation of most PM proteins occurred via a proteasome-independent mechanism. Upregulated proteins included a decoy receptor for TRAIL. Twenty VACV-encoded PM proteins were identified, of which five were not recognised previously as such. Collectively, this dataset constitutes a valuable resource for future studies on antiviral immunity, host-pathogen interaction, poxvirus biology, vector-based vaccine design and oncolytic therapy. Vaccinia virus (VACV) is the vaccine used to eradicate smallpox and an excellent model for studying host-pathogen interactions. Many VACV-mediated immune evasion strategies are known, however how immune cells recognise VACV-infected cells is incompletely understood because of the complexity of surface proteins regulating such interactions. Here, a systematic study of proteins on the cell surface at different times during infection with VACV is presented. This shows not only the precise nature and kinetics of appearance of VACV proteins, but also the selective alteration of cellular surface proteins. The latter thereby identified potential novel immune evasion strategies and host proteins regulating immune activation. Comprehensive comparisons with published datasets provided further insight into mechanisms used to regulate surface protein expression. Such comparisons also identified proteins that are targeted by both VACV and human cytomegalovirus (HCMV), and which are therefore likely to represent host proteins regulating immune recognition and activation. Collectively, this work provides a valuable resource for studying viral immune evasion mechanisms and novel host proteins critical in host immunity.
Collapse
Affiliation(s)
| | | | - Lior Soday
- Cambridge Institute for Medical Research, University of Cambridge, United Kingdom
| | | | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, United Kingdom
| | | | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, United Kingdom
- * E-mail: (MPW); (GLS)
| | - Geoffrey L. Smith
- Department of Pathology, University of Cambridge, United Kingdom
- * E-mail: (MPW); (GLS)
| |
Collapse
|
217
|
Martínez-Martínez E, Atzei P, Vionnet C, Roubaty C, Kaeser-Pebernard S, Naef R, Dengjel J. A Dual-Acting Nitric Oxide Donor and Phosphodiesterase 5 Inhibitor Activates Autophagy in Primary Skin Fibroblasts. Int J Mol Sci 2022; 23:ijms23126860. [PMID: 35743299 PMCID: PMC9224465 DOI: 10.3390/ijms23126860] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/07/2022] [Accepted: 06/17/2022] [Indexed: 02/01/2023] Open
Abstract
Wound healing pathologies are an increasing problem in ageing societies. Chronic, non-healing wounds, which cause high morbidity and severely reduce the quality of life of affected individuals, are frequently observed in aged individuals and people suffering from diseases affected by the Western lifestyle, such as diabetes. Causal treatments that support proper wound healing are still scarce. Here, we performed expression proteomics to study the effects of the small molecule TOP-N53 on primary human skin fibroblasts and keratinocytes. TOP-N53 is a dual-acting nitric oxide donor and phosphodiesterase-5 inhibitor increasing cGMP levels to support proper wound healing. In contrast to keratinocytes, which did not exhibit global proteome alterations, TOP-N53 had profound effects on the proteome of skin fibroblasts. In fibroblasts, TOP-N53 activated the cytoprotective, lysosomal degradation pathway autophagy and induced the expression of the selective autophagy receptor p62/SQSTM1. Thus, activation of autophagy might in part be responsible for beneficial effects of TOP-N53.
Collapse
Affiliation(s)
- Esther Martínez-Martínez
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
| | - Paola Atzei
- Topadur Pharma AG, Grabenstrasse 11A, 8952 Schlieren, Switzerland; (P.A.); (R.N.)
| | - Christine Vionnet
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
| | - Carole Roubaty
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
| | - Stephanie Kaeser-Pebernard
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
| | - Reto Naef
- Topadur Pharma AG, Grabenstrasse 11A, 8952 Schlieren, Switzerland; (P.A.); (R.N.)
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, 1700 Fribourg, Switzerland; (E.M.-M.); (C.V.); (C.R.); (S.K.-P.)
- Correspondence:
| |
Collapse
|
218
|
Protein Markers for the Identification of Cork Oak Plants Infected with Phytophthora cinnamomi by Applying an (α, β)-k-Feature Set Approach. FORESTS 2022. [DOI: 10.3390/f13060940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cork oak decline in Mediterranean forests is a complex phenomenon, observed with remarkable frequency in the southern part of the Iberian Peninsula, causing the weakening and death of these woody plants. The defoliation of the canopy, the presence of dry peripheral branches, and exudations on the trunk are visible symptoms used for the prognosis of decline, complemented by the presence of Phytophthora cinnamomi identified in the rhizosphere of the trees and adjacent soils. Recently, a large proteomic dataset obtained from the leaves of cork oak plants inoculated and non-inoculated with P. cinnamomi has become available. We explored it to search for an optimal set of proteins, markers of the biological pattern of interaction with the oomycete. Thus, using published data from the cork oak leaf proteome, we mathematically modelled the problem as an α, β-k-Feature Set Problem to select molecular markers. A set of proteins (features) that represent dominant effects on the host metabolism resulting from pathogen action on roots was found. These results contribute to an early diagnosis of biochemical changes occurring in cork oak associated with P. cinnamomi infection. We hypothesize that these markers may be decisive in identifying trees that go into decline due to interactions with the pathogen, assisting the management of cork oak forest ecosystems.
Collapse
|
219
|
Stimulating Effect of Trichococcus flocculiformis on a Coculture of Syntrophomonas wolfei and Methanospirillum hungatei. Appl Environ Microbiol 2022; 88:e0039122. [PMID: 35699440 PMCID: PMC9275234 DOI: 10.1128/aem.00391-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Syntrophic anaerobic consortia comprised of fatty acid-degrading bacteria and hydrogen/formate-scavenging methanogenic archaea are of central importance for balanced and resilient natural and manufactured ecosystems: anoxic sediments, soils, and wastewater treatment bioreactors. Previously published studies investigated interaction between the syntrophic bi-cultures, but little information is available on the influence of fermentative bacteria on syntrophic fatty acid oxidation, even though fermentative organisms are always present together with syntrophic partners in the above-mentioned ecosystems. Here, we present experimental observations of stimulated butyrate oxidation and methane generation by a coculture of Syntrophomonas wolfei with any of the following methanogens: Methanospirillum hungatei, Methanobrevibacter arboriphilus, or Methanobacterium formicicum due to the addition of a fermentative Trichococcus flocculiformis strain ES5. The addition of T. flocculiformis ES5 to the syntrophic cocultures led to an increase in the rates of butyrate consumption (120%) and volumetric methane production (150%). Scanning electron microscopy of the most positively affected coculture (S. wolfei, M. hungatei, and T. flocculiformis ES5) revealed a tendency of T. flocculiformis ES5 to aggregate with the syntrophic partners. Analysis of coculture’s proteome with or without addition of the fermentative bacterium points to a potential link with signal transducing systems of M. hungatei, as well as activation of additional butyryl coenzyme A dehydrogenase and an electron transfer flavoprotein in S. wolfei. IMPORTANCE Results from the present study open doors to fascinating research on complex microbial cultures in anaerobic environments (of biotechnological and ecological relevance). Such studies of defined mixed populations are critical to understanding the highly intertwined natural and engineered microbial systems and to developing more reliable and trustable metabolic models. By investigating the existing cultured microbial consortia, like the ones described here, we can acquire knowledge on microbial interactions that go beyond “who feeds whom” relations but yet benefit the parties involved. Transfer of signaling compounds and stimulation of gene expression are examples of indirect influence that members of mixed communities can exert on each other. Understanding such microbial relationships will enable development of new sustainable biotechnologies with mixed microbial cocultures and contribute to the general understanding of the complex natural microbial interactions.
Collapse
|
220
|
Fritsch S, Gasser V, Peukert C, Pinkert L, Kuhn L, Perraud Q, Normant V, Brönstrup M, Schalk IJ. Uptake Mechanisms and Regulatory Responses to MECAM- and DOTAM-Based Artificial Siderophores and Their Antibiotic Conjugates in Pseudomonas aeruginosa. ACS Infect Dis 2022; 8:1134-1146. [PMID: 35500104 DOI: 10.1021/acsinfecdis.2c00049] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The development of new antibiotics against Gram-negative bacteria has to deal with the low permeability of the outer membrane. This obstacle can be overcome by utilizing siderophore-dependent iron uptake pathways as entrance routes for antibiotic uptake. Iron-chelating siderophores are actively imported by bacteria, and their conjugation to antibiotics allows smuggling the latter into bacterial cells. Synthetic siderophore mimetics based on MECAM (1,3,5-N,N',N″-tris-(2,3-dihydroxybenzoyl)-triaminomethylbenzene) and DOTAM (1,4,7,10-tetrakis(carbamoylmethyl)-1,4,7,10-tetraazacyclododecane) cores, both chelating iron via catechol groups, have been recently applied as versatile carriers of functional cargo. In the present study, we show that MECAM and the MECAM-ampicillin conjugate 3 transport iron into Pseudomonas aeruginosa cells via the catechol-type outer membrane transporters PfeA and PirA and DOTAM solely via PirA. Differential proteomics and quantitative real-time polymerase chain reaction (qRT-PCR) showed that MECAM import induced the expression of pfeA, whereas 3 led to an increase in the expression of pfeA and ampc, a gene conferring ampicillin resistance. The presence of DOTAM did not induce the expression of pirA but upregulated the expression of two zinc transporters (cntO and PA0781), pointing out that bacteria become zinc starved in the presence of this compound. Iron uptake experiments with radioactive 55Fe demonstrated that import of this nutrient by MECAM and DOTAM was as efficient as with the natural siderophore enterobactin. The study provides a functional validation for DOTAM- and MECAM-based artificial siderophore mimetics as vehicles for the delivery of cargo into Gram-negative bacteria.
Collapse
Affiliation(s)
- Sarah Fritsch
- CNRS, University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Illkirch, Strasbourg 67070, France
| | - Véronique Gasser
- CNRS, University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Illkirch, Strasbourg 67070, France
| | - Carsten Peukert
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, Braunschweig 38124, Germany
| | - Lukas Pinkert
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, Braunschweig 38124, Germany
| | - Lauriane Kuhn
- Plateforme Proteomique Strasbourg-Esplanade, Institut de Biologie Moléculaire et Cellulaire, CNRS, FR1589, 15 rue Descartes, Strasbourg Cedex F-67084, France
| | - Quentin Perraud
- CNRS, University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Illkirch, Strasbourg 67070, France
| | - Vincent Normant
- CNRS, University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Illkirch, Strasbourg 67070, France
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, Braunschweig 38124, Germany
- German Center for Infection Research (DZIF), Site Hannover-Braunschweig, Braunschweig 38124, Germany
- Center of Biomolecular Drug Research (BMWZ), Leibniz Universität, Hannover 30159, Germany
| | - Isabelle J. Schalk
- CNRS, University of Strasbourg, UMR7242, ESBS, Bld Sébastien Brant, F-67412 Illkirch, Strasbourg 67070, France
| |
Collapse
|
221
|
Grau-Bové X, Navarrete C, Chiva C, Pribasnig T, Antó M, Torruella G, Galindo LJ, Lang BF, Moreira D, López-Garcia P, Ruiz-Trillo I, Schleper C, Sabidó E, Sebé-Pedrós A. A phylogenetic and proteomic reconstruction of eukaryotic chromatin evolution. Nat Ecol Evol 2022; 6:1007-1023. [PMID: 35680998 PMCID: PMC7613034 DOI: 10.1038/s41559-022-01771-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/21/2022] [Indexed: 02/06/2023]
Abstract
Histones and associated chromatin proteins have essential functions in eukaryotic genome organization and regulation. Despite this fundamental role in eukaryotic cell biology, we lack a phylogenetically-comprehensive understanding of chromatin evolution. Here, we combine comparative proteomics and genomics analysis of chromatin in eukaryotes and archaea. Proteomics uncovers the existence of histone post-translational modifications in Archaea. However, archaeal histone modifications are scarce, in contrast with the highly conserved and abundant marks we identify across eukaryotes. Phylogenetic analysis reveals that chromatin-associated catalytic functions (e.g., methyltransferases) have pre-eukaryotic origins, whereas histone mark readers and chaperones are eukaryotic innovations. We show that further chromatin evolution is characterized by expansion of readers, including capture by transposable elements and viruses. Overall, our study infers detailed evolutionary history of eukaryotic chromatin: from its archaeal roots, through the emergence of nucleosome-based regulation in the eukaryotic ancestor, to the diversification of chromatin regulators and their hijacking by genomic parasites.
Collapse
Affiliation(s)
- Xavier Grau-Bové
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Cristina Navarrete
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | | | - Thomas Pribasnig
- Department of Functional and Evolutionary Ecology, Archaea Biology Unit, University of Vienna, Vienna, Austria
| | - Meritxell Antó
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Barcelona, Spain
| | - Guifré Torruella
- Unité d'Ecologie Systématique et Evolution, CNRS, Université Paris-Saclay, AgroParisTech, Orsay, France
| | - Luis Javier Galindo
- Unité d'Ecologie Systématique et Evolution, CNRS, Université Paris-Saclay, AgroParisTech, Orsay, France
| | - Bernd Franz Lang
- Robert Cedergren Centre in Bioinformatics and Genomics, Department of Biochemistry, Université de Montréal, Montréal, Quebec, Canada
| | - David Moreira
- Unité d'Ecologie Systématique et Evolution, CNRS, Université Paris-Saclay, AgroParisTech, Orsay, France
| | - Purificación López-Garcia
- Unité d'Ecologie Systématique et Evolution, CNRS, Université Paris-Saclay, AgroParisTech, Orsay, France
| | - Iñaki Ruiz-Trillo
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Barcelona, Spain.,Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Christa Schleper
- Department of Functional and Evolutionary Ecology, Archaea Biology Unit, University of Vienna, Vienna, Austria
| | - Eduard Sabidó
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Arnau Sebé-Pedrós
- Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain. .,Universitat Pompeu Fabra (UPF), Barcelona, Spain.
| |
Collapse
|
222
|
Lancaster SM, Lee-McMullen B, Abbott CW, Quijada JV, Hornburg D, Park H, Perelman D, Peterson DJ, Tang M, Robinson A, Ahadi S, Contrepois K, Hung CJ, Ashland M, McLaughlin T, Boonyanit A, Horning A, Sonnenburg JL, Snyder MP. Global, distinctive, and personal changes in molecular and microbial profiles by specific fibers in humans. Cell Host Microbe 2022; 30:848-862.e7. [PMID: 35483363 PMCID: PMC9187607 DOI: 10.1016/j.chom.2022.03.036] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/19/2022] [Accepted: 03/25/2022] [Indexed: 12/11/2022]
Abstract
Dietary fibers act through the microbiome to improve cardiovascular health and prevent metabolic disorders and cancer. To understand the health benefits of dietary fiber supplementation, we investigated two popular purified fibers, arabinoxylan (AX) and long-chain inulin (LCI), and a mixture of five fibers. We present multiomic signatures of metabolomics, lipidomics, proteomics, metagenomics, a cytokine panel, and clinical measurements on healthy and insulin-resistant participants. Each fiber is associated with fiber-dependent biochemical and microbial responses. AX consumption associates with a significant reduction in LDL and an increase in bile acids, contributing to its observed cholesterol reduction. LCI is associated with an increase in Bifidobacterium. However, at the highest LCI dose, there is increased inflammation and elevation in the liver enzyme alanine aminotransferase. This study yields insights into the effects of fiber supplementation and the mechanisms behind fiber-induced cholesterol reduction, and it shows effects of individual, purified fibers on the microbiome.
Collapse
Affiliation(s)
- Samuel M Lancaster
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Brittany Lee-McMullen
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Charles Wilbur Abbott
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Jeniffer V Quijada
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Daniel Hornburg
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Heyjun Park
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Dalia Perelman
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Dylan J Peterson
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Michael Tang
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Aaron Robinson
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Sara Ahadi
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Chia-Jui Hung
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Melanie Ashland
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Tracey McLaughlin
- Division of Endocrinology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Anna Boonyanit
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Aaron Horning
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Justin L Sonnenburg
- Department of Microbiology & Immunology, Stanford School of Medicine, Stanford, CA 94305, USA; Chan-Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Michael P Snyder
- Department of Genetics, Stanford School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
223
|
In-depth Profiling and Quantification of the Lysine Acetylome in Hepatocellular Carcinoma with a Trapped Ion Mobility Mass Spectrometer. Mol Cell Proteomics 2022; 21:100255. [PMID: 35688384 PMCID: PMC9294201 DOI: 10.1016/j.mcpro.2022.100255] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 05/30/2022] [Accepted: 06/05/2022] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most common cause of cancer-related death worldwide with limited therapeutic options. Comprehensive investigation of protein posttranslational modifications in HCC is still limited. Lysine acetylation is one of the most common types of posttranslational modification involved in many cellular processes and plays crucial roles in the regulation of cancer. In this study, we analyzed the proteome and K-acetylome in eight pairs of HCC tumors and normal adjacent tissues using a timsTOF Pro instrument. As a result, we identified 9219 K-acetylation sites in 2625 proteins, of which 1003 sites exhibited differential acetylation levels between tumors and normal adjacent tissues. Interestingly, many novel tumor-specific K-acetylation sites were characterized, for example, filamin A (K865), filamin B (K697), and cofilin (K19), suggesting altered activities of these cytoskeleton-modulating molecules, which may contribute to tumor metastasis. In addition, we observed an overall suppression of protein K-acetylation in HCC tumors, especially for enzymes from various metabolic pathways, for example, glycolysis, tricarboxylic acid cycle, and fatty acid metabolism. Moreover, the expression of deacetylase sirtuin 2 (SIRT2) was upregulated in HCC tumors, and its role of deacetylation in HCC cells was further explored by examining the impact of SIRT2 overexpression on the proteome and K-acetylome in Huh7 HCC cells. SIRT2 overexpression reduced K-acetylation of proteins involved in a wide range of cellular processes, including energy metabolism. Furthermore, cellular assays showed that overexpression of SIRT2 in HCC cells inhibited both glycolysis and oxidative phosphorylation. Taken together, our findings provide valuable information to better understand the roles of K-acetylation in HCC and to treat this disease by correcting the aberrant acetylation patterns. K-acetylation was generally reduced in HCC, especially in metabolic enzymes. Deacetylase SIRT2 was upregulated in HCC tumors. SIRT2 overexpression induced broad alteration of protein K-acetylation. SIRT2 overexpression inhibited glycolysis and oxidative phosphorylation.
Collapse
|
224
|
Yan K, Meng Q, He H, Zhu H, Wang Z, Han L, Huang Q, Zhang Z, Yawalkar N, Zhou H, Xu J. iTRAQ-based quantitative proteomics reveals biomarkers/pathways in psoriasis that can predict the efficacy of methotrexate. J Eur Acad Dermatol Venereol 2022; 36:1784-1795. [PMID: 35666151 DOI: 10.1111/jdv.18292] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Methotrexate (MTX) is the first-line medicine to treat psoriasis. So far, there has been less research on protein biomarkers to predict its efficacy by the proteomic technique. OBJECTIVES To evaluate differentially expressed proteins in peripheral mononuclear cells (PBMCs) between good responders (GRs) and non-responders (NRs) after MTX treatment, compared with normal controls (NCs). METHODS We quantified protein expression of PBMCs with 4 GRs and 4 NRs to MTX and 4 NCs by isobaric tags for relative and absolute quantification (iTRAQ), analyzing and identifying proteins related to efficacy of MTX in 18 psoriatic patients. RESULTS A total of 3,177 proteins had quantitative information, and 403 differentially expressed proteins (fold change ≥ 1.2, p < .05) were identified. Compared to NCs, upregulated proteins (ANXA6, RPS27A, EZR, XRCC6), participating in the activation of NF-κB, the JAK-STAT pathway, and neutrophil degranulation were detected in GRs. The proteins (GPV, FN1, STOM), involving platelet activation, signaling and aggregation as well as neutrophil degranulation were significantly downregulated in GRs. These proteins returned to normal levels after MTX treatment. Furthermore, Western blotting identified the expression of ANXA6 and STAT1 in PBMCs, which were significantly downregulated in GRs, but not in NRs. CONCLUSIONS We identified seven differentially expressed and regulated proteins (ANXA6, GPV, FN1, XRCC6, STOM, RPS27A, and EZR) as biomarkers to predict MTX efficacy in NF-κB signaling, JAK-STAT pathways, neutrophil degranulation, platelet activation, signaling and aggregation.
Collapse
Affiliation(s)
- Kexiang Yan
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qian Meng
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Han He
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Hongwen Zhu
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Zhicheng Wang
- Department of Clinical Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Ling Han
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Qiong Huang
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhenghua Zhang
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Nikhil Yawalkar
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Hu Zhou
- CAS Key Laboratory of Receptor Research, Stake Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jinhua Xu
- Institute of Dermatology and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
225
|
Van Meenen J, Leysen H, Chen H, Baccarne R, Walter D, Martin B, Maudsley S. Making Biomedical Sciences publications more accessible for machines. MEDICINE, HEALTH CARE, AND PHILOSOPHY 2022; 25:179-190. [PMID: 35039972 DOI: 10.1007/s11019-022-10069-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/08/2022] [Indexed: 06/14/2023]
Abstract
With the rapidly expanding catalogue of scientific publications, especially within the Biomedical Sciences field, it is becoming increasingly difficult for researchers to search for, read or even interpret emerging scientific findings. PubMed, just one of the current biomedical data repositories, comprises over 33 million citations for biomedical research, and over 2500 publications are added each day. To further strengthen the impact biomedical research, we suggest that there should be more synergy between publications and machines. By bringing machines into the realm of research and publication, we can greatly augment the assessment, investigation and cataloging of the biomedical literary corpus. The effective application of machine-based manuscript assessment and interpretation is now crucial, and potentially stands as the most effective way for researchers to comprehend and process the tsunami of biomedical data and literature. Many biomedical manuscripts are currently published online in poorly searchable document types, with figures and data presented in formats that are partially inaccessible to machine-based approaches. The structure and format of biomedical manuscripts should be adapted to facilitate machine-assisted interrogation of this important literary corpus. In this context, it is important to embrace the concept that biomedical scientists should also write manuscripts that can be read by machines. It is likely that an enhanced human-machine synergy in reading biomedical publications will greatly enhance biomedical data retrieval and reveal novel insights into complex datasets.
Collapse
Affiliation(s)
- Joris Van Meenen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
- Antwerp Research Group for Ocular Science, Department of Translational Neurosciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Hongyu Chen
- Weill Cornell Medical College, New York, NY, USA
| | - Rudi Baccarne
- Anet Library Automation, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Deborah Walter
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Bronwen Martin
- Faculty of Pharmaceutical, Veterinary and Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Lab, Department of Biomedical Sciences, University of Antwerp, Wilrijk, 2610, Antwerp, Belgium.
| |
Collapse
|
226
|
Amaranthus hypochondriacus seeds as a rich source of cysteine rich bioactive peptides. Food Chem 2022; 377:131959. [PMID: 34995961 DOI: 10.1016/j.foodchem.2021.131959] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 12/20/2021] [Accepted: 12/24/2021] [Indexed: 11/23/2022]
Abstract
Amaranthus hypochondriacus is a nutritious alternative grain native to Central and South America. Increased interest in the impact of A. hypochondriacus on the human body has driven characterization of bioactive secondary metabolites. The seeds are known to contain bioactive small molecules but little is known regarding endogenous peptides. Cysteine-rich peptides (CRPs) in foodstuffs are particularly relevant because they are stabilized by disulfide bonds enhancing resistance to digestion. Here, in silico predictions, proteomics, and simulated gastrointestinal digestions are leveraged to identify digestion resistant CRPs within A. hypochondriacus seeds. Thirteen in silico predicted CRPs were detected in a seed extract providing evidence for the translation of five CRP families. Mature forms of six CRPs were characterized via top-down proteomics revealing multiple post-translational modifications. All six peptides demonstrated resistance to simulated gastrointestinal digestion, suggesting that A. hypochondriacus CRPs may exhibit bioactivity after consumption and should be prioritized for further characterization.
Collapse
|
227
|
Muth-Pawlak D, Kreula S, Gollan PJ, Huokko T, Allahverdiyeva Y, Aro EM. Patterning of the Autotrophic, Mixotrophic, and Heterotrophic Proteomes of Oxygen-Evolving Cyanobacterium Synechocystis sp. PCC 6803. Front Microbiol 2022; 13:891895. [PMID: 35694301 PMCID: PMC9175036 DOI: 10.3389/fmicb.2022.891895] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Proteomes of an oxygenic photosynthetic cyanobacterium, Synechocystis sp. PCC 6803, were analyzed under photoautotrophic (low and high CO2, assigned as ATLC and ATHC), photomixotrophic (MT), and light-activated heterotrophic (LAH) conditions. Allocation of proteome mass fraction to seven sub-proteomes and differential expression of individual proteins were analyzed, paying particular attention to photosynthesis and carbon metabolism–centered sub-proteomes affected by the quality and quantity of the carbon source and light regime upon growth. A distinct common feature of the ATHC, MT, and LAH cultures was low abundance of inducible carbon-concentrating mechanisms and photorespiration-related enzymes, independent of the inorganic or organic carbon source. On the other hand, these cells accumulated a respiratory NAD(P)H dehydrogenase I (NDH-11) complex in the thylakoid membrane (TM). Additionally, in glucose-supplemented cultures, a distinct NDH-2 protein, NdbA, accumulated in the TM, while the plasma membrane-localized NdbC and terminal oxidase decreased in abundance in comparison to both AT conditions. Photosynthetic complexes were uniquely depleted under the LAH condition but accumulated under the ATHC condition. The MT proteome displayed several heterotrophic features typical of the LAH proteome, particularly including the high abundance of ribosome as well as amino acid and protein biosynthesis machinery-related components. It is also noteworthy that the two equally light-exposed ATHC and MT cultures allocated similar mass fractions of the total proteome to the seven distinct sub-proteomes. Unique trophic condition-specific expression patterns were likewise observed among individual proteins, including the accumulation of phosphate transporters and polyphosphate polymers storing energy surplus in highly energetic bonds under the MT condition and accumulation under the LAH condition of an enzyme catalyzing cyanophycin biosynthesis. It is concluded that the rigor of cell growth in the MT condition results, to a great extent, by combining photosynthetic activity with high intracellular inorganic carbon conditions created upon glucose breakdown and release of CO2, besides the direct utilization of glucose-derived carbon skeletons for growth. This combination provides the MT cultures with excellent conditions for growth that often exceeds that of mere ATHC.
Collapse
|
228
|
Maffioli E, Angiulli E, Nonnis S, Grassi Scalvini F, Negri A, Tedeschi G, Arisi I, Frabetti F, D’Aniello S, Alleva E, Cioni C, Toni M. Brain Proteome and Behavioural Analysis in Wild Type, BDNF +/- and BDNF -/- Adult Zebrafish ( Danio rerio) Exposed to Two Different Temperatures. Int J Mol Sci 2022; 23:ijms23105606. [PMID: 35628418 PMCID: PMC9146406 DOI: 10.3390/ijms23105606] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Experimental evidence suggests that environmental stress conditions can alter the expression of BDNF and that the expression of this neurotrophin influences behavioural responses in mammalian models. It has been recently demonstrated that exposure to 34 °C for 21 days alters the brain proteome and behaviour in zebrafish. The aim of this work was to investigate the role of BDNF in the nervous system of adult zebrafish under control and heat treatment conditions. For this purpose, zebrafish from three different genotypes (wild type, heterozygous BDNF+/- and knock out BDNF-/-) were kept for 21 days at 26 °C or 34 °C and then euthanized for brain molecular analyses or subjected to behavioural tests (Y-maze test, novel tank test, light and dark test, social preference test, mirror biting test) for assessing behavioural aspects such as boldness, anxiety, social preference, aggressive behaviour, interest for the novel environment and exploration. qRT-PCR analysis showed the reduction of gene expression of BDNF and its receptors after heat treatment in wild type zebrafish. Moreover, proteomic analysis and behavioural tests showed genotype- and temperature-dependent effects on brain proteome and behavioural responding. Overall, the absent expression of BDNF in KO alters (1) the brain proteome by reducing the expression of proteins involved in synapse functioning and neurotransmitter-mediated transduction; (2) the behaviour, which can be interpreted as bolder and less anxious and (3) the cellular and behavioural response to thermal treatment.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
| | - Elisa Angiulli
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University, Via Alfonso Borelli 50, 00161 Rome, Italy; (E.A.); (C.C.)
| | - Simona Nonnis
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
- CRC I-WE (Coordinating Research Centre: Innovation for Well-Being and Environment), University of Milan, 20134 Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
| | - Armando Negri
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (A.N.); (G.T.)
- CRC I-WE (Coordinating Research Centre: Innovation for Well-Being and Environment), University of Milan, 20134 Milan, Italy
| | - Ivan Arisi
- Bioinformatics Facility, European Brain Research Institute (EBRI) “Rita Levi-Montalcini”, 00161 Rome, Italy;
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), 00131 Rome, Italy
| | - Flavia Frabetti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40136 Bologna, Italy;
| | - Salvatore D’Aniello
- Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn Napoli, Villa Comunale, 80121 Napoli, Italy;
| | - Enrico Alleva
- Center for Behavioural Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Carla Cioni
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University, Via Alfonso Borelli 50, 00161 Rome, Italy; (E.A.); (C.C.)
| | - Mattia Toni
- Department of Biology and Biotechnology “Charles Darwin”, Sapienza University, Via Alfonso Borelli 50, 00161 Rome, Italy; (E.A.); (C.C.)
- Correspondence:
| |
Collapse
|
229
|
Uldry AC, Maciel-Dominguez A, Jornod M, Buchs N, Braga-Lagache S, Brodard J, Jankovic J, Bonadies N, Heller M. Effect of Sample Transportation on the Proteome of Human Circulating Blood Extracellular Vesicles. Int J Mol Sci 2022; 23:ijms23094515. [PMID: 35562906 PMCID: PMC9099550 DOI: 10.3390/ijms23094515] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/08/2022] [Accepted: 04/09/2022] [Indexed: 02/01/2023] Open
Abstract
Circulating extracellular vesicles (cEV) are released by many kinds of cells and play an important role in cellular communication, signaling, inflammation modulation, coagulation, and tumor growth. cEV are of growing interest, not only as biomarkers, but also as potential treatment targets. However, very little is known about the effect of transporting biological samples from the clinical ward to the diagnostic laboratory, notably on the protein composition. Pneumatic tube systems (PTS) and human carriers (C) are both routinely used for transport, subjecting the samples to different ranges of mechanical forces. We therefore investigated qualitatively and quantitatively the effect of transport by C and PTS on the human cEV proteome and particle size distribution. We found that samples transported by PTS were subjected to intense, irregular, and multidirectional shocks, while those that were transported by C mostly underwent oscillations at a ground frequency of approximately 4 Hz. PTS resulted in the broadening of nanoparticle size distribution in platelet-free (PFP) but not in platelet-poor plasma (PPP). Cell-type specific cEV-associated protein abundances remained largely unaffected by the transport type. Since residual material of lymphocytes, monocytes, and platelets seemed to dominate cEV proteomes in PPP, it was concluded that PFP should be preferred for any further analyses. Differential expression showed that the impact of the transport method on cEV-associated protein composition was heterogeneous and likely donor-specific. Correlation analysis was nonetheless able to detect that vibration dose, shocks, and imparted energy were associated with different terms depending on the transport, namely in C with cytoskeleton-regulated cell organization activity, and in PTS with a release of extracellular vesicles, mainly from organelle origin, and specifically from mitochondrial structures. Feature selection algorithm identified proteins which, when considered together with the correlated protein-protein interaction network, could be viewed as surrogates of network clusters.
Collapse
Affiliation(s)
- Anne-Christine Uldry
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (A.-C.U.); (A.M.-D.); (M.J.); (N.B.); (S.B.-L.)
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland;
| | - Anabel Maciel-Dominguez
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (A.-C.U.); (A.M.-D.); (M.J.); (N.B.); (S.B.-L.)
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland;
| | - Maïwenn Jornod
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (A.-C.U.); (A.M.-D.); (M.J.); (N.B.); (S.B.-L.)
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland;
| | - Natasha Buchs
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (A.-C.U.); (A.M.-D.); (M.J.); (N.B.); (S.B.-L.)
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland;
| | - Sophie Braga-Lagache
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (A.-C.U.); (A.M.-D.); (M.J.); (N.B.); (S.B.-L.)
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland;
| | - Justine Brodard
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (J.B.); (J.J.)
| | - Jovana Jankovic
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (J.B.); (J.J.)
| | - Nicolas Bonadies
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland;
- Department of Hematology and Central Hematology Laboratory, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland; (J.B.); (J.J.)
| | - Manfred Heller
- Proteomics and Mass Spectrometry Core Facility, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (A.-C.U.); (A.M.-D.); (M.J.); (N.B.); (S.B.-L.)
- Department for BioMedical Research, University of Bern, 3008 Bern, Switzerland;
- Correspondence: ; Tel.: +41-31-684-04-82
| |
Collapse
|
230
|
Föll MC, Volkmann V, Enderle-Ammour K, Timme S, Wilhelm K, Guo D, Vitek O, Bronsert P, Schilling O. Moving translational mass spectrometry imaging towards transparent and reproducible data analyses: a case study of an urothelial cancer cohort analyzed in the Galaxy framework. Clin Proteomics 2022; 19:8. [PMID: 35439943 PMCID: PMC9016955 DOI: 10.1186/s12014-022-09347-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 04/04/2022] [Indexed: 11/24/2022] Open
Abstract
Background Mass spectrometry imaging (MSI) derives spatial molecular distribution maps directly from clinical tissue specimens and thus bears great potential for assisting pathologists with diagnostic decisions or personalized treatments. Unfortunately, progress in translational MSI is often hindered by insufficient quality control and lack of reproducible data analysis. Raw data and analysis scripts are rarely publicly shared. Here, we demonstrate the application of the Galaxy MSI tool set for the reproducible analysis of a urothelial carcinoma dataset. Methods Tryptic peptides were imaged in a cohort of 39 formalin-fixed, paraffin-embedded human urothelial cancer tissue cores with a MALDI-TOF/TOF device. The complete data analysis was performed in a fully transparent and reproducible manner on the European Galaxy Server. Annotations of tumor and stroma were performed by a pathologist and transferred to the MSI data to allow for supervised classifications of tumor vs. stroma tissue areas as well as for muscle-infiltrating and non-muscle infiltrating urothelial carcinomas. For putative peptide identifications, m/z features were matched to the MSiMass list. Results Rigorous quality control in combination with careful pre-processing enabled reduction of m/z shifts and intensity batch effects. High classification accuracy was found for both, tumor vs. stroma and muscle-infiltrating vs. non-muscle infiltrating urothelial tumors. Some of the most discriminative m/z features for each condition could be assigned a putative identity: stromal tissue was characterized by collagen peptides and tumor tissue by histone peptides. Immunohistochemistry confirmed an increased histone H2A abundance in the tumor compared to the stroma tissues. The muscle-infiltration status was distinguished via MSI by peptides from intermediate filaments such as cytokeratin 7 in non-muscle infiltrating carcinomas and vimentin in muscle-infiltrating urothelial carcinomas, which was confirmed by immunohistochemistry. To make the study fully reproducible and to advocate the criteria of FAIR (findability, accessibility, interoperability, and reusability) research data, we share the raw data, spectra annotations as well as all Galaxy histories and workflows. Data are available via ProteomeXchange with identifier PXD026459 and Galaxy results via https://github.com/foellmelanie/Bladder_MSI_Manuscript_Galaxy_links. Conclusion Here, we show that translational MSI data analysis in a fully transparent and reproducible manner is possible and we would like to encourage the community to join our efforts.
Collapse
Affiliation(s)
- Melanie Christine Föll
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115a, 79106, FreiburgFreiburg, Germany. .,Khoury College of Computer Sciences, Northeastern University, Boston, USA.
| | - Veronika Volkmann
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115a, 79106, FreiburgFreiburg, Germany
| | - Kathrin Enderle-Ammour
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115a, 79106, FreiburgFreiburg, Germany
| | - Sylvia Timme
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115a, 79106, FreiburgFreiburg, Germany.,Core Facility for Histopathology and Digital Pathology, Faculty of Medicine, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Konrad Wilhelm
- Department of Urology, Center for Surgery, Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Dan Guo
- Khoury College of Computer Sciences, Northeastern University, Boston, USA
| | - Olga Vitek
- Khoury College of Computer Sciences, Northeastern University, Boston, USA
| | - Peter Bronsert
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115a, 79106, FreiburgFreiburg, Germany.,German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany.,Tumorbank Comprehensive Cancer Center Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Faculty of Medicine, Institute for Surgical Pathology, Medical Center - University of Freiburg, Breisacher Straße 115a, 79106, FreiburgFreiburg, Germany.,German Cancer Consortium (DKTK) and Cancer Research Center (DKFZ), Freiburg, Germany
| |
Collapse
|
231
|
Wörtz J, Smith V, Fallmann J, König S, Thuraisingam T, Walther P, Urlaub H, Stadler PF, Allers T, Hille F, Marchfelder A. Cas1 and Fen1 Display Equivalent Functions During Archaeal DNA Repair. Front Microbiol 2022; 13:822304. [PMID: 35495653 PMCID: PMC9051519 DOI: 10.3389/fmicb.2022.822304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
CRISPR-Cas constitutes an adaptive prokaryotic defence system against invasive nucleic acids like viruses and plasmids. Beyond their role in immunity, CRISPR-Cas systems have been shown to closely interact with components of cellular DNA repair pathways, either by regulating their expression or via direct protein-protein contact and enzymatic activity. The integrase Cas1 is usually involved in the adaptation phase of CRISPR-Cas immunity but an additional role in cellular DNA repair pathways has been proposed previously. Here, we analysed the capacity of an archaeal Cas1 from Haloferax volcanii to act upon DNA damage induced by oxidative stress and found that a deletion of the cas1 gene led to reduced survival rates following stress induction. In addition, our results indicate that Cas1 is directly involved in DNA repair as the enzymatically active site of the protein is crucial for growth under oxidative conditions. Based on biochemical assays, we propose a mechanism by which Cas1 plays a similar function to DNA repair protein Fen1 by cleaving branched intermediate structures. The present study broadens our understanding of the functional link between CRISPR-Cas immunity and DNA repair by demonstrating that Cas1 and Fen1 display equivalent roles during archaeal DNA damage repair.
Collapse
Affiliation(s)
| | - Victoria Smith
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Jörg Fallmann
- Department of Computer Science, Bioinformatics Group, Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
| | - Sabine König
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | | | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, Ulm, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Institute of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Peter F. Stadler
- Department of Computer Science, Bioinformatics Group, Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany
- German Centre for Integrative Biodiversity Research (iDiv) Halle-Jena-Leipzig, Leipzig, Germany
- Competence Center for Scalable Data Services and Solutions, Leipzig Research Center for Civilization Diseases, University Leipzig, Leipzig, Germany
- Facultad de Ciencias, Universidad Nacional de Colombia, Bogotá, Colombia
- Institute for Theoretical Chemistry, University of Vienna, Vienna, Austria
- Center for RNA in Technology and Health, University of Copenhagen, Copenhagen, Denmark
- Santa Fe Institute, Santa Fe, NM, United States
- Max Planck Institute for Mathematics in the Sciences, Leipzig, Germany
| | - Thorsten Allers
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | | | - Anita Marchfelder
- Biology II, Ulm University, Ulm, Germany
- *Correspondence: Anita Marchfelder,
| |
Collapse
|
232
|
Hochstrasser R, Michaelis S, Brülisauer S, Sura T, Fan M, Maaß S, Becher D, Hilbi H. Migration of Acanthamoeba through Legionella biofilms is regulated by the bacterial Lqs-LvbR network, effector proteins and the flagellum. Environ Microbiol 2022; 24:3672-3692. [PMID: 35415862 PMCID: PMC9544456 DOI: 10.1111/1462-2920.16008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 11/30/2022]
Abstract
The environmental bacterium Legionella pneumophila causes the pneumonia Legionnaires' disease. The opportunistic pathogen forms biofilms and employs the Icm/Dot type IV secretion system (T4SS) to replicate in amoebae and macrophages. A regulatory network comprising the Legionella quorum sensing (Lqs) system and the transcription factor LvbR controls bacterial motility, virulence and biofilm architecture. Here we show by comparative proteomics that in biofilms formed by the L. pneumophila ΔlqsR or ΔlvbR regulatory mutants the abundance of proteins encoded by a genomic ‘fitness island’, metabolic enzymes, effector proteins and flagellar components (e.g. FlaA) varies. ∆lqsR or ∆flaA mutants form ‘patchy’ biofilms like the parental strain JR32, while ∆lvbR forms a ‘mat‐like’ biofilm. Acanthamoeba castellanii amoebae migrated more slowly through biofilms of L. pneumophila lacking lqsR, lvbR, flaA, a functional Icm/Dot T4SS (∆icmT), or secreted effector proteins. Clusters of bacteria decorated amoebae in JR32, ∆lvbR or ∆icmT biofilms but not in ∆lqsR or ∆flaA biofilms. The amoeba‐adherent bacteria induced promoters implicated in motility (PflaA) or virulence (PsidC, PralF). Taken together, the Lqs‐LvbR network (quorum sensing), FlaA (motility) and the Icm/Dot T4SS (virulence) regulate migration of A. castellanii through L. pneumophila biofilms, and – apart from the T4SS – govern bacterial cluster formation on the amoebae.
Collapse
Affiliation(s)
- Ramon Hochstrasser
- Institute of Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006, Zürich, Switzerland
| | - Sarah Michaelis
- Institute of Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006, Zürich, Switzerland
| | - Sabrina Brülisauer
- Institute of Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006, Zürich, Switzerland
| | - Thomas Sura
- Institute of Microbiology, University of Greifswald, Felix-Hausdorff-Strasse 8, 17489, Greifswald, Germany
| | - Mingzhen Fan
- Institute of Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006, Zürich, Switzerland
| | - Sandra Maaß
- Institute of Microbiology, University of Greifswald, Felix-Hausdorff-Strasse 8, 17489, Greifswald, Germany
| | - Dörte Becher
- Institute of Microbiology, University of Greifswald, Felix-Hausdorff-Strasse 8, 17489, Greifswald, Germany
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zürich, Gloriastrasse 30, 8006, Zürich, Switzerland
| |
Collapse
|
233
|
A Monoclonal Anti-HMGB1 Antibody Attenuates Neurodegeneration in an Experimental Animal Model of Glaucoma. Int J Mol Sci 2022; 23:ijms23084107. [PMID: 35456925 PMCID: PMC9028318 DOI: 10.3390/ijms23084107] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/02/2022] [Accepted: 04/05/2022] [Indexed: 02/06/2023] Open
Abstract
Neuroinflammation is a crucial process for the loss of retinal ganglion cells (RGC), a major characteristic of glaucoma. High expression of high-mobility group box protein 1 (HMGB1) plays a detrimental role in inflammatory processes and is elevated in the retinas of glaucoma patients. Therefore, this study aimed to investigate the effects of the intravitreal injection of an anti-HMGB1 monoclonal antibody (anti-HMGB1 Ab) in an experimental animal model of glaucoma. Two groups of Spraque Dawley rats received episcleral vein occlusion to chronically elevate intraocular pressure (IOP): (1) the IgG group, intravitreal injection of an unspecific IgG as a control, n = 5, and (2) the HMGB1 group, intravitreal injection of an anti-HMGB1 Ab, n = 6. IOP, retinal nerve fiber layer thickness (RNFLT), and the retinal flash response were monitored longitudinally. Post-mortem examinations included immunohistochemistry, microarray, and mass spectrometric analysis. RNFLT was significantly increased in the HMGB1 group compared with the IgG group (p < 0.001). RGC density showed improved neuronal cell survival in the retina in HMGB1 compared with the IgG group (p < 0.01). Mass spectrometric proteomic analysis of retinal tissue showed an increased abundance of RNA metabolism-associated heterogeneous nuclear ribonucleoproteins (hnRNPs), such as hnRNP U, D, and H2, in animals injected with the anti-HMGB1 Ab, indicating that the application of the antibody may cause increased gene expression. Microarray analysis showed a significantly decreased expression of C-X-C motif chemokine ligand 8 (CXCL8, p < 0.05) and connective tissue growth factor (CTGF, p < 0.01) in the HMGB1 group. Thus, these data suggest that intravitreal injection of anti-HMGB1 Ab reduced HMGB1-dependent inflammatory signaling and mediated RGC neuroprotection.
Collapse
|
234
|
Sui X, Cox D, Nie S, Reid GE, Hatters DM. A Census of Hsp70-Mediated Proteome Solubility Changes upon Recovery from Heat Stress. J Proteome Res 2022; 21:1251-1261. [PMID: 35388693 DOI: 10.1021/acs.jproteome.1c00920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Eukaryotic cells respond to heat shock through several regulatory processes including upregulation of stress responsive chaperones and reversible shutdown of cellular activities through formation of protein assemblies. However, the underlying regulatory mechanisms of the recovery of these heat-induced protein assemblies remain largely elusive. Here, we measured the proteome abundance and solubility changes during recovery from heat shock in the mouse Neuro2a cell line. We found that prefoldins and translation machinery are rapidly down-regulated as the first step in the heat shock response. Analysis of proteome solubility reveals that a rapid mobilization of protein quality control machineries, along with changes in cellular energy metabolism, translational activity, and actin cytoskeleton are fundamental to the early stress responses. In contrast, longer term adaptation to stress involves renewal of core cellular components. Inhibition of the Hsp70 family, pivotal for the heat shock response, selectively and negatively affects the ribosomal machinery and delays the solubility recovery of many nuclear proteins. ProteomeXchange: PXD030069.
Collapse
Affiliation(s)
- Xiaojing Sui
- Department of Molecular Biosciences, Rice Institute for Biomedical Research, Northwestern University, Evanston, Illinois 60208, United States
| | - Dezerae Cox
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Shuai Nie
- Melbourne Mass Spectrometry and Proteomics Facility, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Gavin E Reid
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia.,School of Chemistry, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Danny M Hatters
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
235
|
Liu Y, Charamis N, Boeren S, Blok J, Lewis AG, Smid EJ, Abee T. Physiological Roles of Short-Chain and Long-Chain Menaquinones (Vitamin K2) in Lactococcus cremoris. Front Microbiol 2022; 13:823623. [PMID: 35369466 PMCID: PMC8965153 DOI: 10.3389/fmicb.2022.823623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 01/17/2022] [Indexed: 01/07/2023] Open
Abstract
Lactococcus cremoris and L. lactis are well known for their occurrence and applications in dairy fermentations, but their niche extends to a range of natural and food production environments. L. cremoris and L. lactis produce MKs (vitamin K2), mainly as the long-chain forms represented by MK-9 and MK-8, and a detectable number of short-chain forms represented by MK-3. The physiological significance of the different MK forms in the lifestyle of these bacterial species has not been investigated extensively. In this study, we used L. cremoris MG1363 to construct mutants producing different MK profiles by deletion of genes encoding (i) a menaquinone-specific isochorismate synthase, (ii) a geranyltranstransferase, and (iii) a prenyl diphosphate synthase. These gene deletions resulted in (i) a non-MK producer (ΔmenF), (ii) a presumed MK-1 producer (ΔispA), and (iii) an MK-3 producer (Δllmg_0196), respectively. By examining the phenotypes of the MG1363 wildtype strain and respective mutants, including biomass accumulation, stationary phase survival, oxygen consumption, primary metabolites, azo dye/copper reduction, and proteomes, under aerobic, anaerobic, and respiration-permissive conditions, we could infer that short-chain MKs like MK-1 and MK-3 are preferred to mediate extracellular electron transfer and reaction with extracellular oxygen, while the long-chain MKs like MK-9 and MK-8 are more efficient in aerobic respiratory electron transport chain. The different electron transfer routes mediated by short-chain and long-chain MKs likely support growth and survival of L. cremoris in a range of (transiently) anaerobic and aerobic niches including food fermentations, highlighting the physiological significance of diverse MKs in L. cremoris.
Collapse
Affiliation(s)
- Yue Liu
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | - Nikolaos Charamis
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University and Research, Wageningen, Netherlands
| | - Joost Blok
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | | | - Eddy J Smid
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| | - Tjakko Abee
- Food Microbiology, Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
236
|
Vanuopadath M, Raveendran D, Nair BG, Nair SS. Venomics and antivenomics of Indian spectacled cobra (Naja naja) from the Western Ghats. Acta Trop 2022; 228:106324. [PMID: 35093326 DOI: 10.1016/j.actatropica.2022.106324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 01/03/2023]
Abstract
Venom proteome profiling of Naja naja from the Western Ghats region in Kerala was achieved through SDS-PAGE and RP-HPLC followed by Q-TOF LC-MS/MS analysis, incorporating PEAKS and Novor assisted de novo sequencing methodologies. A total of 115 proteins distributed across 17 different enzymatic and non-enzymatic venom protein families were identified through conventional and 39 peptides through homology-driven proteomics approaches. Fourteen peptides derived through de novo complements the Mascot data indicating the importance of homology-driven approaches in improving protein sequence information. Among the protein families identified, glutathione peroxidase and endonuclease were reported for the first time in the Indian cobra venom. Immunological cross-reactivity assessed using Indian polyvalent antivenoms suggested that VINS showed better EC50 (2.48 µg/mL) value than that of PSAV (6.04 µg/mL) and Virchow (6.03 µg/mL) antivenoms. Western blotting experiments indicated that all the antivenoms elicited poor binding specificities, especially towards low molecular mass proteins. Second-generation antivenomics studies revealed that VINS antivenom was less efficient to detect many low molecular mass proteins such as three-finger toxins and Kunitz-type serine protease Inhibitors. Taken together, the present study enabled a large-scale characterization of the venom proteome of Naja naja from the Western Ghats and emphasized the need for developing more efficient antivenoms.
Collapse
Affiliation(s)
| | - Dileepkumar Raveendran
- Indriyam Biologics Pvt. Ltd., SCTIMST-TIMed, BMT Wing-Poojappura, Thiruvananthapuram 695 012, Kerala, India
| | | | | |
Collapse
|
237
|
Gupta R, Min CW, Park SR, Kim ST. Label-free proteome data of susceptible and resistant rice cultivars in response to Xanthomonas oryzae pv. oryzae inoculation. Data Brief 2022; 41:107890. [PMID: 35198671 PMCID: PMC8842008 DOI: 10.1016/j.dib.2022.107890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 11/24/2022] Open
Abstract
Here we report the data associated with the article: "Comparative proteome profiling of susceptible and resistant rice cultivars identified an arginase involved in rice defense against Xanthomonas oryzae pv. oryzae" [1]. Bacterial blight disease caused by Xanthomonas oryzae pv. oryzae (Xoo) is one of the most devastating diseases of rice across the globe; however, the underlying molecular mechanism of rice-Xoo interaction is currently not well understood. In this manuscript, we report the proteome profiles of rice leaves generated using a label-free quantitative proteomic analysis using QExactive™ Orbitrap High-Resolution Mass Spectrometer, MapMan, and rice interactome viewer [1].
Collapse
Affiliation(s)
- Ravi Gupta
- College of General Education, Kookmin University, Seoul 02707, Republic of Korea
| | - Cheol Woo Min
- Department of Plant Bioscience, Life and Industry Convergence Research Institute, Pusan National University, Miryang 627-707, Republic of Korea
| | - Sang-Ryeol Park
- National Institute of Agricultural Sciences, Rural Development Administration, Jeonju 54874, Republic of Korea
| | - Sun Tae Kim
- Department of Plant Bioscience, Life and Industry Convergence Research Institute, Pusan National University, Miryang 627-707, Republic of Korea
| |
Collapse
|
238
|
Sidhanth C, Bindhya S, Krishnapriya S, Manasa P, Shabna A, Alifia J, Patole C, Kumar V, Garg M, Ganesan TS. Phosphoproteome of signaling by ErbB2 in ovarian cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140768. [PMID: 35158093 DOI: 10.1016/j.bbapap.2022.140768] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/07/2022] [Accepted: 02/04/2022] [Indexed: 06/14/2023]
Abstract
The gene for receptor tyrosine kinase ErbB2 is amplified in breast and ovarian tumours. The linear pathway by which signals are transduced through ErbB2 are well known. However, second generation questions that address spatial aspects of signaling remain. To address this, we have undertaken a mass spectrometry approach to identify phosphoproteins specific for ErbB2 using the inhibitors Lapatinib and CP724714 in ovarian cancer cells. The ErbB2 specific proteins identified in SKOV-3 cells were Myristoylated alanine-rich C-kinase substrate, Protein capicua homolog, Protein peptidyl isomerase G, Protein PRRC2C, Chromobox homolog1 and PRP4 homolog. We have evaluated three phosphoproteins PKM2, Aldose reductase and MARCKS in SKOV-3 cells. We observed that PKM2 was phosphorylated by EGF but was not inhibited by Lapatinib and CP724714. The activity of aldose reductase in reducing NADPH as a substrate was significantly higher in EGF stimulated cells which was inhibited by Lapatinib and CP724714 but not by Geftinib (EGFR inhibitor). MARCKS was phosphorylated on stimulation of SKOV-3 cells with EGF that was inhibited by Lapatinib and CP724714 which was dependent on the kinase activity of ErbB2. These results have identified phosphoproteins that are specific to ErbB2 which have not been previously reported and sets the basis for future experiments.
Collapse
Affiliation(s)
- C Sidhanth
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - S Bindhya
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - S Krishnapriya
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - P Manasa
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - A Shabna
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India
| | - J Alifia
- Mass Spectrometry Facility Proteomics, National Centre for Biological Sciences (NCBS), Bangalore, India
| | - C Patole
- Mass Spectrometry Facility Proteomics, National Centre for Biological Sciences (NCBS), Bangalore, India
| | - V Kumar
- Mass Spectrometry and Proteomics Core Facility, University of Nebraska Medical Center, Omaha, NE, USA
| | - M Garg
- Amity Institute of Molecular Medicine & Stem cell Research, Amity University, Delhi, India
| | - T S Ganesan
- Laboratory for Cancer Biology, Departments of Medical Oncology and Clinical Research, Cancer Institute (WIA), Chennai, India.
| |
Collapse
|
239
|
Bense S, Witte J, Preuße M, Koska M, Pezoldt L, Dröge A, Hartmann O, Müsken M, Schulze J, Fiebig T, Bähre H, Felgner S, Pich A, Häussler S. Pseudomonas aeruginosa post-translational responses to elevated c-di-GMP levels. Mol Microbiol 2022; 117:1213-1226. [PMID: 35362616 DOI: 10.1111/mmi.14902] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 11/29/2022]
Abstract
C-di-GMP signaling can directly influence bacterial behavior by affecting the functionality of c-di-GMP-binding proteins. In addition, c-di-GMP can exert a global effect on gene transcription or translation, e.g., via riboswitches or by binding to transcription factors. In this study, we investigated the effects of changes in intracellular c-di-GMP levels on gene expression and protein production in the opportunistic pathogen Pseudomonas aeruginosa. We induced c-di-GMP production via an ectopically introduced diguanylate cyclase and recorded the transcriptional, translational as well as proteomic profile of the cells. We demonstrate that rising levels of c-di-GMP under growth conditions otherwise characterized by low c-di-GMP levels caused a switch to a non-motile, auto-aggregative P. aeruginosa phenotype. This phenotypic switch became apparent before any c-di-GMP-dependent role on transcription, translation, or protein abundance was observed. Our results suggest that rising global c-di-GMP pools first affects the motility phenotype of P. aeruginosa by altering protein functionality and only then global gene transcription.
Collapse
Affiliation(s)
- Sarina Bense
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute for Molecular Bacteriology, TWINCORE GmbH, Center of Clinical and Experimental Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Center for Infection Research, Hannover, Germany
| | - Julius Witte
- Institute for Molecular Bacteriology, TWINCORE GmbH, Center of Clinical and Experimental Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Center for Infection Research, Hannover, Germany.,Research Core Unit Proteomics and Institute for Toxicology, Hannover Medical School, Hannover, Germany
| | - Matthias Preuße
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Michal Koska
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute for Molecular Bacteriology, TWINCORE GmbH, Center of Clinical and Experimental Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Center for Infection Research, Hannover, Germany
| | - Lorena Pezoldt
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute for Molecular Bacteriology, TWINCORE GmbH, Center of Clinical and Experimental Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Center for Infection Research, Hannover, Germany
| | - Astrid Dröge
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Oliver Hartmann
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute for Molecular Bacteriology, TWINCORE GmbH, Center of Clinical and Experimental Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Center for Infection Research, Hannover, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Center for Infection Research, Braunschweig, Germany
| | - Julia Schulze
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Timm Fiebig
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Heike Bähre
- Research Core Unit Metabolomics and Institute of Pharmacology, Hannover Medical School, Hannover, Germany. Infection Research, Hannover, Germany
| | - Sebastian Felgner
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute for Molecular Bacteriology, TWINCORE GmbH, Center of Clinical and Experimental Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Center for Infection Research, Hannover, Germany
| | - Andreas Pich
- Research Core Unit Proteomics and Institute for Toxicology, Hannover Medical School, Hannover, Germany
| | - Susanne Häussler
- Department of Molecular Bacteriology, Helmholtz Center for Infection Research, Braunschweig, Germany.,Institute for Molecular Bacteriology, TWINCORE GmbH, Center of Clinical and Experimental Infection Research, a joint venture of the Hannover Medical School and the Helmholtz Center for Infection Research, Hannover, Germany.,Department of Clinical Microbiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark.,Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
240
|
Hulme CH, Fuller HR, Riddell J, Shirran SL, Botting CH, Osman A, Wright KT. Investigation of the blood proteome in response to spinal cord injury in rodent models. Spinal Cord 2022; 60:320-325. [PMID: 34601498 PMCID: PMC8989679 DOI: 10.1038/s41393-021-00692-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 11/13/2022]
Abstract
STUDY DESIGN Explanatory and mechanistic study. OBJECTIVES A better understanding of the 'whole-body' response following spinal cord injury (SCI) is needed to guide future research aimed at developing novel therapeutic interventions and identifying prognostic indicators for SCI. This study aimed to characterise the blood proteome following contusion or complete SCI compared to a sham injury in rat models. SETTING United Kingdom. METHODS Pooled blood samples from one and seven days after a contusion (serum; n = 5) or from 14 days and 112 days post-complete transection SCI (plasma; n = 8) and their sham-injured counterparts were subjected to independent iTRAQ nanoflow liquid chromatography tandem mass-spectrometry proteomic analyses. Pathway analyses of the proteins that were differentially abundant between SCI and their matched sham injured counterparts were completed to indicate biological pathways that may be changed in response to SCI. RESULTS Eleven and 42 proteins were differentially abundant (≥±2.0 FC; p ≤ 0.05) between the contusion SCI and sham injured animals at 24 h and seven days post-injury, respectively. Seven and tweleve proteins were differentially abundant between complete and sham injured rats at 14 and 112 days post-injury, respectively. Acute-phase response signalling and Liver X Receptor/Retinoic X Receptor activation were identified as differentially regulated pathways in both models of SCI. CONCLUSIONS We have utilised longitudinal preclinical SCI models to provide an insight into the blood proteome changes that result following SCI and to highlight a number of biological pathways of interest for future studies.
Collapse
Affiliation(s)
- Charlotte H Hulme
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, UK
- Midlands Centre for Spinal Injuries, RJAH Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Heidi R Fuller
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, UK
| | - John Riddell
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow, UK
| | - Sally L Shirran
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, North Haugh, St Andrews, UK
| | - Catherine H Botting
- BSRC Mass Spectrometry and Proteomics Facility, University of St Andrews, North Haugh, St Andrews, UK
| | - Aheed Osman
- Midlands Centre for Spinal Injuries, RJAH Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Karina T Wright
- School of Pharmacy and Bioengineering, Keele University, Keele, Staffordshire, UK.
- Midlands Centre for Spinal Injuries, RJAH Orthopaedic Hospital, Oswestry, Shropshire, UK.
| |
Collapse
|
241
|
Dozio E, Maffioli E, Vianello E, Nonnis S, Grassi Scalvini F, Spatola L, Roccabianca P, Tedeschi G, Corsi Romanelli MM. A Wide-Proteome Analysis to Identify Molecular Pathways Involved in Kidney Response to High-Fat Diet in Mice. Int J Mol Sci 2022; 23:ijms23073809. [PMID: 35409168 PMCID: PMC8999052 DOI: 10.3390/ijms23073809] [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: 03/03/2022] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/18/2022] Open
Abstract
The etiopathogenesis of obesity-related chronic kidney disease (CKD) is still scarcely understood. To this aim, we assessed the effect of high-fat diet (HF) on molecular pathways leading to organ damage, steatosis, and fibrosis. Six-week-old male C57BL/6N mice were fed HF diet or normal chow for 20 weeks. Kidneys were collected for genomic, proteomic, histological studies, and lipid quantification. The main findings were as follows: (1) HF diet activated specific pathways leading to fibrosis and increased fatty acid metabolism; (2) HF diet promoted a metabolic shift of lipid metabolism from peroxisomes to mitochondria; (3) no signs of lipid accumulation and/or fibrosis were observed, histologically; (4) the early signs of kidney damage seemed to be related to changes in membrane protein expression; (5) the proto-oncogene MYC was one of the upstream transcriptional regulators of changes occurring in protein expression. These results demonstrated the potential usefulness of specific selected molecules as early markers of renal injury in HF, while histomorphological changes become visible later in obesity-related CDK. The integration of these information with data from biological fluids could help the identification of biomarkers useful for the early detection and prevention of tissue damage in clinical practice.
Collapse
Affiliation(s)
- Elena Dozio
- Laboratory of Clinical Pathology, Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.D.); (M.M.C.R.)
| | - Elisa Maffioli
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
| | - Elena Vianello
- Laboratory of Clinical Pathology, Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.D.); (M.M.C.R.)
- Correspondence: ; Tel.: +39-02-50315342
| | - Simona Nonnis
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
- CRC “Innovation for Well-Being and Environment” (I-WE), Università degli Studi di Milano, 29133 Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
| | - Leonardo Spatola
- Division of Nephrology, Dialysis and Renal Transplantation, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy;
| | - Paola Roccabianca
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
| | - Gabriella Tedeschi
- Department of Veterinary Medicine and Animal Science, Università degli Studi di Milano, 26900 Lodi, Italy; (E.M.); (S.N.); (F.G.S.); (P.R.); (G.T.)
- CRC “Innovation for Well-Being and Environment” (I-WE), Università degli Studi di Milano, 29133 Milan, Italy
| | - Massimiliano Marco Corsi Romanelli
- Laboratory of Clinical Pathology, Department of Biomedical Sciences for Health, Università degli Studi di Milano, 20133 Milan, Italy; (E.D.); (M.M.C.R.)
- Service of Laboratory Medicine1-Clinical Pathology, IRCCS Policlinico San Donato, 20097 San Donato Milanese, Italy
| |
Collapse
|
242
|
Ceballos-Laita L, Takahashi D, Uemura M, Abadía J, López-Millán AF, Rodríguez-Celma J. Effects of Fe and Mn Deficiencies on the Root Protein Profiles of Tomato ( Solanum lycopersicum) Using Two-Dimensional Electrophoresis and Label-Free Shotgun Analyses. Int J Mol Sci 2022; 23:ijms23073719. [PMID: 35409079 PMCID: PMC8998858 DOI: 10.3390/ijms23073719] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 12/04/2022] Open
Abstract
Iron (Fe) and manganese (Mn) are two essential elements for plants that compete for the same uptake transporters and show conflicting interactions at the regulatory level. In order to understand the differential response to both metal deficiencies in plants, two proteomic techniques (two-dimensional gel electrophoresis and label-free shotgun) were used to study the proteome profiles of roots from tomato plants grown under Fe or Mn deficiency. A total of 119 proteins changing in relative abundance were confidently quantified and identified, including 35 and 91 in the cases of Fe deficiency and Mn deficiency, respectively, with 7 of them changing in both deficiencies. The identified proteins were categorized according to function, and GO-enrichment analysis was performed. Data showed that both deficiencies provoked a common and intense cell wall remodelling. However, the response observed for Fe and Mn deficiencies differed greatly in relation to oxidative stress, coumarin production, protein, nitrogen, and energy metabolism.
Collapse
Affiliation(s)
- Laura Ceballos-Laita
- Plant Stress Physiology Group, Plant Nutrition Department, Aula Dei Experimental Station, CSIC, 50059 Zaragoza, Spain; (L.C.-L.); (J.A.); (A.F.L.-M.)
| | - Daisuke Takahashi
- United Graduate School of Agricultural Sciences, Iwate University, Morioka 020-8550, Japan; (D.T.); (M.U.)
- Department of Plant-Bioscience, Faculty of Agriculture, Iwate University, Morioka 020-8550, Japan
| | - Matsuo Uemura
- United Graduate School of Agricultural Sciences, Iwate University, Morioka 020-8550, Japan; (D.T.); (M.U.)
- Department of Plant-Bioscience, Faculty of Agriculture, Iwate University, Morioka 020-8550, Japan
| | - Javier Abadía
- Plant Stress Physiology Group, Plant Nutrition Department, Aula Dei Experimental Station, CSIC, 50059 Zaragoza, Spain; (L.C.-L.); (J.A.); (A.F.L.-M.)
| | - Ana Flor López-Millán
- Plant Stress Physiology Group, Plant Nutrition Department, Aula Dei Experimental Station, CSIC, 50059 Zaragoza, Spain; (L.C.-L.); (J.A.); (A.F.L.-M.)
| | - Jorge Rodríguez-Celma
- Plant Stress Physiology Group, Plant Nutrition Department, Aula Dei Experimental Station, CSIC, 50059 Zaragoza, Spain; (L.C.-L.); (J.A.); (A.F.L.-M.)
- Correspondence:
| |
Collapse
|
243
|
Chalise U, Daseke MJ, Kalusche WJ, Konfrst SR, Rodriguez-Paar JR, Flynn ER, Cook LM, Becirovic-Agic M, Lindsey ML. Macrophages secrete murinoglobulin-1 and galectin-3 to regulate neutrophil degranulation after myocardial infarction. Mol Omics 2022; 18:186-195. [PMID: 35230372 PMCID: PMC8963000 DOI: 10.1039/d1mo00519g] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/11/2022] [Indexed: 02/03/2023]
Abstract
Inflammation presides early after myocardial infarction (MI) as a key event in cardiac wound healing. Ischemic cardiomyocytes secrete inflammatory cues to stimulate infiltration of leukocytes, predominantly macrophages and neutrophils. Infiltrating neutrophils degranulate to release a series of proteases including matrix metalloproteinase (MMP)-9 to break down extracellular matrix and remove necrotic myocytes to create space for the infarct scar to form. While neutrophil to macrophage communication has been explored, the reverse has been understudied. We used a proteomics approach to catalogue the macrophage secretome at MI day 1. Murinoglobulin-1 (MUG1) was the highest-ranked secreted protein (4.1-fold upregulated at MI day 1 vs. day 0 pre-MI cardiac macrophages, p = 0.004). By transcriptomics evaluation, galectin-3 (Lgals3) was 2.2-fold upregulated (p = 0.008) in MI day 1 macrophages. We explored the direct roles of MUG1 and Lgals3 on neutrophil degranulation. MUG1 blunted while Lgals3 amplified neutrophil degranulation in response to phorbol 12-myristate 13-acetate or interleukin-1β, as measured by MMP-9 secretion. Lgals3 itself also stimulated MMP-9 secretion. To determine if MUG1 regulated Lgals3, we co-stimulated neutrophils with MUG1 and Lgals3. MUG1 limited degranulation stimulated by Lgals3 by 64% (p < 0.001). In vivo, MUG1 was elevated in the infarct region at MI days 1 and 3, while Lgals3 increased at MI day 7. The ratio of MUG1 to Lgals3 positively correlated with infarct wall thickness, revealing that MUG1 attenuated infarct wall thinning. In conclusion, macrophages at MI day 1 secrete MUG1 to limit and Lgals3 to accentuate neutrophil degranulation to regulate infarct wall thinning.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Michael J Daseke
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - William J Kalusche
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Shelby R Konfrst
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Jocelyn R Rodriguez-Paar
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Elizabeth R Flynn
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mediha Becirovic-Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, USA
| |
Collapse
|
244
|
Weiß L, Gaelings L, Reiner T, Mergner J, Kuster B, Fehér A, Hensel G, Gahrtz M, Kumlehn J, Engelhardt S, Hückelhoven R. Posttranslational modification of the RHO of plants protein RACB by phosphorylation and cross-kingdom conserved ubiquitination. PLoS One 2022; 17:e0258924. [PMID: 35333858 PMCID: PMC8956194 DOI: 10.1371/journal.pone.0258924] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/10/2021] [Indexed: 11/19/2022] Open
Abstract
Small RHO-type G-proteins act as signaling hubs and master regulators of polarity in eukaryotic cells. Their activity is tightly controlled, as defective RHO signaling leads to aberrant growth and developmental defects. Two major processes regulate G-protein activity: canonical shuttling between different nucleotide bound states and posttranslational modification (PTM), of which the latter can support or suppress RHO signaling, depending on the individual PTM. In plants, regulation of Rho of plants (ROPs) signaling activity has been shown to act through nucleotide exchange and GTP hydrolysis, as well as through lipid modification, but there is little data available on phosphorylation or ubiquitination of ROPs. Hence, we applied proteomic analyses to identify PTMs of the barley ROP RACB. We observed in vitro phosphorylation by barley ROP binding kinase 1 and in vivo ubiquitination of RACB. Comparative analyses of the newly identified RACB phosphosites and human RHO protein phosphosites revealed conservation of modified amino acid residues, but no overlap of actual phosphorylation patterns. However, the identified RACB ubiquitination site is conserved in all ROPs from Hordeum vulgare, Arabidopsis thaliana and Oryza sativa and in mammalian Rac1 and Rac3. Point mutation of this ubiquitination site leads to stabilization of RACB. Hence, this highly conserved lysine residue may regulate protein stability across different kingdoms.
Collapse
Affiliation(s)
- Lukas Weiß
- Chair of Phytopathology, Technical University of Munich (TUM), Freising, Germany
| | - Lana Gaelings
- Chair of Phytopathology, Technical University of Munich (TUM), Freising, Germany
| | - Tina Reiner
- Chair of Phytopathology, Technical University of Munich (TUM), Freising, Germany
| | - Julia Mergner
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
- Bavarian Biomolecular Mass Spectrometry Center (BayBioMS), TUM, Freising, Germany
| | - Attila Fehér
- Chair of Plant Biology, University of Szeged, and Institute of Plant Biology, Biological Research Centre, Szeged, Hungary
| | - Götz Hensel
- Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Manfred Gahrtz
- Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Jochen Kumlehn
- Institute of Plant Genetics and Crop Plant Research (IPK), Gatersleben, Germany
| | - Stefan Engelhardt
- Chair of Phytopathology, Technical University of Munich (TUM), Freising, Germany
| | - Ralph Hückelhoven
- Chair of Phytopathology, Technical University of Munich (TUM), Freising, Germany
| |
Collapse
|
245
|
Graham RLJ, McMullen AA, Moore G, Dempsey-Hibbert NC, Myers B, Graham C. SWATH-MS identification of CXCL7, LBP, TGFβ1 and PDGFRβ as novel biomarkers in human systemic mastocytosis. Sci Rep 2022; 12:5087. [PMID: 35332176 PMCID: PMC8948255 DOI: 10.1038/s41598-022-08345-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022] Open
Abstract
Mastocytosis is a rare myeloproliferative disease, characterised by accumulation of neoplastic mast cells in one or several organs. It presents as cutaneous or systemic. Patients with advanced systemic mastocytosis have a median survival of 3.5 years. The aetiology of mastocytosis is poorly understood, patients present with a broad spectrum of varying clinical symptoms that lack specificity to point clearly to a definitive diagnosis. Discovery of novel blood borne biomarkers would provide a tractable method for rapid identification of mastocytosis and its sub-types. Moving towards this goal, we carried out a clinical biomarker study on blood from twenty individuals (systemic mastocytosis: n = 12, controls: n = 8), which were subjected to global proteome investigation using the novel technology SWATH-MS. This identified several putative biomarkers for systemic mastocytosis. Orthogonal validation of these putative biomarkers was achieved using ELISAs. Utilising this workflow, we identified and validated CXCL7, LBP, TGFβ1 and PDGF receptor-β as novel biomarkers for systemic mastocytosis. We demonstrate that CXCL7 correlates with neutrophil count offering a new insight into the increased prevalence of anaphylaxis in mastocytosis patients. Additionally, demonstrating the utility of SWATH-MS for the discovery of novel biomarkers in the systemic mastocytosis diagnostic sphere.
Collapse
Affiliation(s)
- R L J Graham
- School of Biological Sciences, Queens University Belfast, Chlorine Gardens, Belfast, BT9 5DL, UK
| | - A A McMullen
- Department of Life Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK
| | - G Moore
- School of Biological Sciences, Queens University Belfast, Chlorine Gardens, Belfast, BT9 5DL, UK
| | - N C Dempsey-Hibbert
- Department of Life Sciences, Manchester Metropolitan University, Manchester, M1 5GD, UK
| | - B Myers
- University Hospitals of Leicester NHS Trust, Leicester, LE3 9QP, UK
| | - C Graham
- School of Biological Sciences, Queens University Belfast, Chlorine Gardens, Belfast, BT9 5DL, UK.
| |
Collapse
|
246
|
Gu H, Yang J, Zhang J, Song Y, Zhang Y, Xu P, Zhu Y, Wang L, Zhang P, Li L, Chen D, Sun Q. PCBP2 maintains antiviral signaling homeostasis by regulating cGAS enzymatic activity via antagonizing its condensation. Nat Commun 2022; 13:1564. [PMID: 35322803 PMCID: PMC8943206 DOI: 10.1038/s41467-022-29266-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/07/2022] [Indexed: 02/07/2023] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) plays a major role in detecting pathogenic DNA. It produces cyclic dinucleotide cGAMP, which subsequently binds to the adaptor protein STING and further triggers antiviral innate immune responses. However, the molecular mechanisms regulating cGAS enzyme activity remain largely unknown. Here, we characterize the cGAS-interacting protein Poly(rC)-binding protein 2 (PCBP2), which plays an important role in controlling cGAS enzyme activity, thereby mediating appropriate cGAS-STING signaling transduction. We find that PCBP2 overexpression reduces cGAS-STING antiviral signaling, whereas loss of PCBP2 significantly increases cGAS activity. Mechanistically, we show that PCBP2 negatively regulates anti-DNA viral signaling by specifically interacting with cGAS but not other components. Moreover, PCBP2 decreases cGAS enzyme activity by antagonizing cGAS condensation, thus ensuring the appropriate production of cGAMP and balancing cGAS-STING signal transduction. Collectively, our findings provide insight into how the cGAS-mediated antiviral signaling is regulated.
Collapse
Affiliation(s)
- Haiyan Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing, 100101, China.,Institute of Biomedical Research, Yunnan University, Kunming, 650500, China.,Institute of Stem Cells and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.,School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Yang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing, 100101, China
| | - Jiayu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing, 100101, China.,Institute of Stem Cells and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.,School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Song
- Institute of Biomedical Research, Yunnan University, Kunming, 650500, China
| | - Yao Zhang
- Institute of Biomedical Research, Yunnan University, Kunming, 650500, China
| | - Pengfei Xu
- Institute of Biomedical Research, Yunnan University, Kunming, 650500, China
| | - Yuanxiang Zhu
- Institute of Biomedical Research, Yunnan University, Kunming, 650500, China
| | - Liangliang Wang
- Institute of Biomedical Research, Yunnan University, Kunming, 650500, China
| | - Pengfei Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing, 100101, China
| | - Lin Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing, 100101, China.,Institute of Stem Cells and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Dahua Chen
- Institute of Biomedical Research, Yunnan University, Kunming, 650500, China.
| | - Qinmiao Sun
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Datun Road, Chaoyang District, Beijing, 100101, China. .,Institute of Stem Cells and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China. .,School of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
247
|
Identification and characterisation of sPEPs in Cryptococcus neoformans. Fungal Genet Biol 2022; 160:103688. [PMID: 35339703 DOI: 10.1016/j.fgb.2022.103688] [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: 01/04/2022] [Revised: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 11/24/2022]
Abstract
Short open reading frame (sORF)-encoded peptides (sPEPs) have been found across a wide range of genomic locations in a variety of species. To date, their identification, validation, and characterisation in the human fungal pathogen Cryptococcus neoformans has been limited due to a lack of standardised protocols. We have developed an enrichment process that enables sPEP detection within a protein sample from this polysaccharide-encapsulated yeast, and implemented proteogenomics to provide insights into the validity of predicted and hypothetical sORFs annotated in the C. neoformans genome. Novel sORFs were discovered within the 5' and 3' UTRs of known transcripts as well as in "non-coding" RNAs. One novel candidate, dubbed NPB1, that resided in an RNA annotated as "non-coding", was chosen for characterisation. Through the creation of both specific point mutations and a full deletion allele, the function of the new sPEP, Npb1, was shown to resemble that of the bacterial trans-translation protein SmpB.
Collapse
|
248
|
Jahangir S, John P, Bhatti A, Aslam MM, Mehmood Malik J, Anderson JR, Peffers MJ. LC-MS/MS-Based Serum Protein Profiling for Identification of Candidate Biomarkers in Pakistani Rheumatoid Arthritis Patients. Life (Basel) 2022; 12:life12030464. [PMID: 35330214 PMCID: PMC8955720 DOI: 10.3390/life12030464] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis is an autoimmune disorder of complex disease etiology. Currently available serological diagnostic markers lack in terms of sensitivity and specificity and thus additional biomarkers are warranted for early disease diagnosis and management. We aimed to screen and compare serum proteome profiles of rheumatoid arthritis serotypes with healthy controls in the Pakistani population for identification of potential disease biomarkers. Serum samples from rheumatoid arthritis patients and healthy controls were enriched for low abundance proteins using ProteoMinerTM columns. Rheumatoid arthritis patients were assigned to one of the four serotypes based on anti-citrullinated peptide antibodies and rheumatoid factor. Serum protein profiles were analyzed via liquid chromatography-tandem mass spectrometry. The changes in the protein abundances were determined using label-free quantification software ProgenesisQITM followed by pathway analysis. Findings were validated in an independent cohort of patients and healthy controls using an enzyme-linked immunosorbent assay. A total of 213 proteins were identified. Comparative analysis of all groups (false discovery rate < 0.05, >2-fold change, and identified with ≥2 unique peptides) identified ten proteins that were differentially expressed between rheumatoid arthritis serotypes and healthy controls including pregnancy zone protein, selenoprotein P, C4b-binding protein beta chain, apolipoprotein M, N-acetylmuramoyl-L-alanine amidase, catalytic chain, oncoprotein-induced transcript 3 protein, Carboxypeptidase N subunit 2, Apolipoprotein C-I and Apolipoprotein C-III. Pathway analysis predicted inhibition of liver X receptor/retinoid X receptor activation pathway and production of nitric oxide and reactive oxygen species pathway in macrophages in all serotypes. A catalogue of potential serum biomarkers for rheumatoid arthritis were identified. These biomarkers can be further evaluated in larger cohorts from different populations for their diagnostic and prognostic potential.
Collapse
Affiliation(s)
- Sidrah Jahangir
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan; (S.J.); (A.B.)
| | - Peter John
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan; (S.J.); (A.B.)
- Correspondence: ; Tel.: +92-051-9085-6151
| | - Attya Bhatti
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad 44000, Pakistan; (S.J.); (A.B.)
| | - Muhammad Muaaz Aslam
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA 15216, USA;
| | | | - James R. Anderson
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK; (J.R.A.); (M.J.P.)
| | - Mandy J. Peffers
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool L7 8TX, UK; (J.R.A.); (M.J.P.)
| |
Collapse
|
249
|
Pantasis S, Friemel J, Brütsch SM, Hu Z, Krautbauer S, Liebisch G, Dengjel J, Weber A, Werner S, Bordoli MR. Vertebrate lonesome kinase modulates the hepatocyte secretome to prevent perivascular liver fibrosis and inflammation. J Cell Sci 2022; 135:275016. [PMID: 35293576 PMCID: PMC9016620 DOI: 10.1242/jcs.259243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 03/08/2022] [Indexed: 11/20/2022] Open
Abstract
Vertebrate lonesome kinase (VLK) is the only known extracellular tyrosine kinase, but its physiological functions are largely unknown. We show that VLK is highly expressed in hepatocytes of neonatal mice, but downregulated during adulthood. To determine the role of VLK in liver homeostasis and regeneration, we generated mice with a hepatocyte-specific knockout of the VLK gene (Pkdcc). Cultured progenitor cells established from primary hepatocytes of Pkdcc knockout mice produced a secretome, which promoted their own proliferation in 3D spheroids and proliferation of cultured fibroblasts. In vivo, Pkdcc knockout mice developed liver steatosis with signs of inflammation and perivascular fibrosis upon aging, combined with expansion of liver progenitor cells. In response to chronic CCl4-induced liver injury, the pattern of deposited collagen was significantly altered in these mice. The liver injury marker alpha-fetoprotein (AFP) was increased in the secretome of VLK-deficient cultured progenitor cells and in liver tissues of aged or CCl4-treated knockout mice. These results support a key role for VLK and extracellular protein phosphorylation in liver homeostasis and repair through paracrine control of liver cell function and regulation of appropriate collagen deposition. This article has an associated First Person interview with the first author of the paper. Summary: The secreted protein kinase VLK is released from hepatocytes and protects the liver from perivascular fibrosis and inflammation.
Collapse
Affiliation(s)
- Sophia Pantasis
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology ETH, Otto-Stern Weg 7, CH-8093, Zurich, Switzerland
| | - Juliane Friemel
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Salome Mirjam Brütsch
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology ETH, Otto-Stern Weg 7, CH-8093, Zurich, Switzerland
| | - Zehan Hu
- Department of Biology, Université de Fribourg, Chemin du Musée 10, CH-1700, Fribourg, Switzerland
| | - Sabrina Krautbauer
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany
| | - Joern Dengjel
- Department of Biology, Université de Fribourg, Chemin du Musée 10, CH-1700, Fribourg, Switzerland
| | - Achim Weber
- Department of Pathology and Molecular Pathology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology ETH, Otto-Stern Weg 7, CH-8093, Zurich, Switzerland
| | - Mattia Renato Bordoli
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology ETH, Otto-Stern Weg 7, CH-8093, Zurich, Switzerland
| |
Collapse
|
250
|
Skariyachan S, Taskeen N, Kishore AP, Krishna BV. Recent advances in plastic degradation - From microbial consortia-based methods to data sciences and computational biology driven approaches. JOURNAL OF HAZARDOUS MATERIALS 2022; 426:128086. [PMID: 34933258 DOI: 10.1016/j.jhazmat.2021.128086] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 12/11/2021] [Accepted: 12/13/2021] [Indexed: 06/14/2023]
Abstract
The conventional methods of plastic waste management such as mechanical and chemical recycling, landfill complemented by incineration and pyrosis have limited scope. Thus, microbiological-based approaches by the application of microbial consortia or cocultures are appropriate, cost-effective, and eco-friendly to manage plastic wastes. Screening of novel plastic degrading microorganisms, the formulation of microbial consortia, and utilisation of their enzymes probably play a role in plastic waste management. The by-products of microbial degradation of plastic waste can be used as bio-energy sources, that aids in the development of cost-effective bio-digesters. The recent advancements in computational biology and bioinformatics play a vital role in understanding the molecular basis of enzymatic degradation of plastic polymers by microorganisms. Understanding the three-dimensional structures of plastic degrading enzymes and their metabolic pathways play a vital role in studying the microbial degradation of plastics. The present review highlights the scope of various microorganisms and their enzymes in plastic degradation. The review emphasizes the applications of co-cultures or microbial consortia-based approaches for the enhanced degradation of plastic polymers and the production of value-added end products that can be used as the prototypes of bioenergy sources. The review also provides a comprehensive outlook on the applications of data sciences, computational biology, and bioinformatics resources, and web-based tools towards the study of microbial degradation of plastic polymers.
Collapse
Affiliation(s)
| | - Neha Taskeen
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, Karnataka, Pin 560078, India
| | - Alice Preethi Kishore
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, Karnataka, Pin 560078, India
| | - Bhavya Venkata Krishna
- Department of Biotechnology, Dayananda Sagar College of Engineering, Bangalore, Karnataka, Pin 560078, India
| |
Collapse
|