1
|
Doye A, Chaintreuil P, Verhoeyen E, Boyer L. [RAC2 gain-of-function mutations causing immunodeficiency drive activation of the NLRP3 inflammasome]. Med Sci (Paris) 2025; 41:133-135. [PMID: 40028950 DOI: 10.1051/medsci/2025008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Affiliation(s)
- Anne Doye
- Université Côte d'Azur, Inserm U1065, Centre méditerranéen de médecine moléculaire, Nice, France
| | - Paul Chaintreuil
- Université Côte d'Azur, Inserm U1065, Centre méditerranéen de médecine moléculaire, Nice, France
| | - Els Verhoeyen
- Université Côte d'Azur, Inserm U1065, Centre méditerranéen de médecine moléculaire, Nice, France
| | - Laurent Boyer
- Université Côte d'Azur, Inserm U1065, Centre méditerranéen de médecine moléculaire, Nice, France
| |
Collapse
|
2
|
Malik HS, Bliska JB. Guards and decoys: RIPoptosome and inflammasome pathway regulators of bacterial effector-triggered immunity. PLoS Pathog 2025; 21:e1012884. [PMID: 39883598 PMCID: PMC11781737 DOI: 10.1371/journal.ppat.1012884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025] Open
Abstract
Virulent microbes produce proteins that interact with host cell targets to promote pathogenesis. For example, virulent bacterial pathogens have proteins called effectors that are typically enzymes and are secreted into host cells. To detect and respond to the activities of effectors, diverse phyla of host organisms evolved effector-triggered immunity (ETI). In ETI, effectors are often sensed indirectly by detection of their virulence activities in host cells. ETI mechanisms can be complex and involve several classes of host proteins. Guards monitor the functional or physical integrity of another host protein, the guardee or decoy, and become activated to initiate an immune response when the guardee or decoy is modified or disrupted by an effector. A guardee typically has an intrinsic anti-pathogen function and is the intended target of an effector. A decoy structurally mimics a host protein that has intrinsic anti-pathogen activity and is unintentionally targeted by an effector. A decoy can be an individual protein, or a protein domain integrated into a guard. Here, we review the origins of ETI and focus on 5 mechanisms, in which the key steps of a pathway can include activation of a caspase by a RIPoptosome or inflammasome, formation of pores in the plasma membrane, release of cytokines and ending in cell death by pyroptosis. Survey of the 5 mechanisms, which have been shown to be host protective in mouse models of bacterial infection, reveal how distinct regulators of RIPoptosome or inflammasome pathways can act as guards or integrated decoys to trigger ETI. Common themes are highlighted and the limited mechanistic understanding of ETI bactericidal activity is discussed.
Collapse
Affiliation(s)
- Haleema Sadia Malik
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| | - James B. Bliska
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, United States of America
| |
Collapse
|
3
|
Doye A, Chaintreuil P, Lagresle-Peyrou C, Batistic L, Marion V, Munro P, Loubatier C, Chirara R, Sorel N, Bessot B, Bronnec P, Contenti J, Courjon J, Giordanengo V, Jacquel A, Barbry P, Couralet M, Aladjidi N, Fischer A, Cavazzana M, Mallebranche C, Visvikis O, Kracker S, Moshous D, Verhoeyen E, Boyer L. RAC2 gain-of-function variants causing inborn error of immunity drive NLRP3 inflammasome activation. J Exp Med 2024; 221:e20231562. [PMID: 39212656 PMCID: PMC11363864 DOI: 10.1084/jem.20231562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 02/15/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
A growing number of patients presenting severe combined immunodeficiencies attributed to monoallelic RAC2 variants have been identified. The expression of the RHO GTPase RAC2 is restricted to the hematopoietic lineage. RAC2 variants have been described to cause immunodeficiencies associated with high frequency of infection, leukopenia, and autoinflammatory features. Here, we show that specific RAC2 activating mutations induce the NLRP3 inflammasome activation leading to the secretion of IL-1β and IL-18 from macrophages. This activation depends on the activation state of the RAC2 variant and is mediated by the downstream kinase PAK1. Inhibiting the RAC2-PAK1-NLRP3 inflammasome pathway might be considered as a potential treatment for these patients.
Collapse
Affiliation(s)
- Anne Doye
- Université Côte d’Azur, INSERM, C3M, Nice, France
| | | | - Chantal Lagresle-Peyrou
- Université Paris Cité, Paris, France
- Imagine Institute, INSERM UMR 1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | | | | | | | | | - Rayana Chirara
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Nataël Sorel
- Imagine Institute, INSERM UMR 1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Boris Bessot
- Imagine Institute, INSERM UMR 1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Pauline Bronnec
- Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Lyon, France
- Université Claude Bernard Lyon 1, CNRS UMR5308, École normale supérieure de Lyon, Lyon, France
| | - Julie Contenti
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Johan Courjon
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Nice, France
| | - Valerie Giordanengo
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Université Côte d’Azur, Centre Hospitalier Universitaire de Nice, Nice, France
| | | | - Pascal Barbry
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia-Antipolis, France
| | - Marie Couralet
- Université Côte d’Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Sophia-Antipolis, France
| | - Nathalie Aladjidi
- Centre de Référence National des Cytopénies Autoimmunes de l’Enfant, Pediatric Hematologic Unit, Centre d’Investigation Clinique Plurithématique INSERM 1401, University Hospital of Bordeaux, Bordeaux, France
| | - Alain Fischer
- Imagine Institute, INSERM UMR 1163, Paris, France
- Necker Hospital, Pediatric Hematology-Immunology and Rheumatology Unit, Assistance Publique-Hôpitaux de Paris, Paris, France
- Collège de France, Paris, France
| | - Marina Cavazzana
- Université Paris Cité, Paris, France
- Imagine Institute, INSERM UMR 1163, Paris, France
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM, Paris, France
| | - Coralie Mallebranche
- Université d’Angers, Université de Nantes, Inserm, CNRS, CRCI2NA, SFR ICAT, Angers, France
- Centre Hospitalier Universitaire Angers, Pediatric Immuno-Hemato-Oncology Unit, Angers, France
| | | | - Sven Kracker
- Université Paris Cité, Paris, France
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM UMR 1163, Paris, France
| | - Despina Moshous
- Laboratoire Dynamique du Génome et Système Immunitaire, Imagine Institute, INSERM UMR 1163, Paris, France
- Centre de Référence des Déficits Immunitaires Héréditaires, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Université Paris Cité, Paris, France
| | - Els Verhoeyen
- Université Côte d’Azur, INSERM, C3M, Nice, France
- Centre International de Recherche en Infectiologie, Université de Lyon, Inserm U1111, Lyon, France
| | | |
Collapse
|
4
|
Wang H, Yung MM, Xuan Y, Chen F, Chan W, Siu MK, Long R, Jia S, Liang Y, Xu D, Song Z, Tsui SK, Ngan HY, Chan KK, Chan DW. Polyunsaturated fatty acids promote M2-like TAM deposition via dampening RhoA-YAP1 signaling in the ovarian cancer microenvironment. Exp Hematol Oncol 2024; 13:90. [PMID: 39198883 PMCID: PMC11360340 DOI: 10.1186/s40164-024-00558-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Peritoneal metastases frequently occur in epithelial ovarian cancer (EOC), resulting in poor prognosis and survival rates. Tumor-associated-macrophages (TAMs) massively infiltrate into ascites spheroids and are multi-polarized as protumoral M2-like phenotype, orchestrating the immunosuppression and promoting tumor progression. However, the impact of omental conditioned medium/ascites (OCM/AS) on TAM polarization and its function in tumor progression remains elusive. METHODS The distribution and polarization of TAMs in primary and omental metastatic EOC patients' tumors and ascites were examined by m-IHC, FACS analysis, and immunofluorescence. QPCR, immunofluorescence, FACS analysis, lipid staining assay, ROS assay, and Seahorse real-time cell metabolic assay characterized TAMs as being polarized in the ascites microenvironment. The oncogenic role of TAMs in tumor cells was demonstrated by co-cultured migration/invasion, proliferation, and spheroid formation assays. Mechanistic studies of the regulations of TAM polarization were performed by using RNA-Seq, GTPase pull-down, G-LISA activation assays, and other biochemical assays. A Yap1 macrophages (MФs) conditional knockout (cKO) mouse model demonstrated the roles of YAP1 in TAM polarization status and its pro-metastatic function. Finally, the anti-metastatic potential of targeting TAMs through restoring YAP1 by pharmacological agonist XMU MP1 was demonstrated in vitro and in vivo. RESULTS Abundant polyunsaturated fatty acids (PUFAs) in OCM/AS suppressed RhoA-GTPase activities, which, in turn, downregulated nuclear YAP1 in MФs, leading to increased protumoral TAM polarization accompanied by elevated OXPHOS metabolism. Abolishment of YAP1 in MФs further confirmed that a higher M2/M1 ratio of TAM polarization could alleviate CD8+ T cell infiltration and cytotoxicity in vivo. Consistently, the loss of YAP1 has been observed in EOC metastatic tissues, suggesting its clinical relevance. On the contrary, restoration of YAP1 expression by pharmaceutical inhibition of MST1/2 induced conversion of M2-to-M1-like polarized MФs, elevating the infiltration of CD8+ T cells and attenuating tumor growth. CONCLUSION This study revealed that PUFAs-enriched OCM/AS of EOC promotes M2-like TAM polarization through RhoA-YAP1 inhibition, where YAP1 downregulation is required for accelerating protumoral M2-like TAM polarization, thereby causing immunosuppression and enhancing tumor progression. Conversion of M2-to-M1-like polarized MФs through Yap1 activation inhibits tumor progression and contributes to developing potential TAMs-targeted immunotherapies in combating EOC peritoneal metastases.
Collapse
Affiliation(s)
- Huogang Wang
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang, P.R. China
| | - Mingo Mh Yung
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Yang Xuan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Fushun Chen
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Waisun Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Michelle Ky Siu
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Runying Long
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Shuo Jia
- Eye Center, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, P.R. China
| | - Yonghao Liang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Dakang Xu
- Faculty of Medical Laboratory Science, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, P.R. China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, Zhejiang, P.R. China
| | - Stephen Kw Tsui
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China
| | - Hextan Ys Ngan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China
| | - Karen Kl Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China.
| | - David W Chan
- Department of Obstetrics & Gynaecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, P.R. China.
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, P.R. China.
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, Guangdong, P.R. China.
| |
Collapse
|
5
|
Paria P, Chakraborty HJ, Pakhira A, Devi MS, Das Mohapatra PK, Behera BK. Identification of virulence-associated factors in Vibrio parahaemolyticus with special reference to moonlighting protein: a secretomics study. Int Microbiol 2024; 27:765-779. [PMID: 37702858 DOI: 10.1007/s10123-023-00429-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/14/2023]
Abstract
Vibrio parahaemolyticus causes seafood-borne gastroenteritis infection in human which can even lead to death. The pathogenic strain of V. parahaemolyticus secretes different types of virulence factors that are directly injected into the host cell by a different type of secretion system which helps bacteria to establish its own ecological niche within the organism. Therefore, the aim of this study was to isolate the extracellular secreted proteins from the trh positive strain of V. parahaemolyticus and identify them using two-dimensional gel electrophoresis and MALDI-TOFMS/MS. Seventeen different cellular proteins viz, Carbamoyl-phosphate synthase, 5-methyltetrahydropteroyltriglutamate, tRNA-dihydrouridine synthase, Glycerol-3-phosphate dehydrogenase, Orotidine 5'-phosphate decarboxylase, Molybdenum import ATP-binding protein, DnaJ, DNA polymerase IV, Ribosomal RNA small subunit methyltransferase G, ATP synthase subunit delta and gamma, Ribosome-recycling factor, 4-hydroxy-3-methylbut-2-en-1-yl diphosphate synthase, tRNA pseudouridine synthase B, Ditrans, polycis-undecaprenyl-diphosphate synthase, Oxygen-dependent coproporphyrinogen-III oxidase, and Peptide deformylase 2 were identified which are mainly involved in different metabolic and biosynthetic pathways. Furthermore, the molecular function of the identified proteins were associated with catalytic activity, ligase activity, transporter, metal binding, and ATP synthase when they are intercellular. However, to understand the importance of these secreted proteins in the infection and survival of bacteria inside the host cell, pathogen-host protein-protein interactions (PPIs) were carried out which identified the association of eight secreted proteins with 41 human proteins involved in different cellular pathways, including ubiquitination degradation, adhesion, inflammation, immunity, and programmed cell death. The present study provides unreported strategies on host-cell environment's survival and adaptation mechanisms for the successful establishment of infections and intracellular propagation.
Collapse
Affiliation(s)
- Prasenjit Paria
- Aquatic Environmental Biotechnology and Nanotechnology Division, ICAR-Central Inland Fisheries Research Institute, Barrackpore, Kolkata, West Bengal, 700120, India
- Vidyasagar University, Midnapur, West Bengal, 721102, India
| | - Hirak Jyoti Chakraborty
- Aquatic Environmental Biotechnology and Nanotechnology Division, ICAR-Central Inland Fisheries Research Institute, Barrackpore, Kolkata, West Bengal, 700120, India
| | - Abhijit Pakhira
- Department of Zoology, Vivekananda Mahavidyalaya, Hooghly, West Bengal, 712405, India
| | - Manoharmayum Shaya Devi
- Aquatic Environmental Biotechnology and Nanotechnology Division, ICAR-Central Inland Fisheries Research Institute, Barrackpore, Kolkata, West Bengal, 700120, India
| | | | - Bijay Kumar Behera
- Aquatic Environmental Biotechnology and Nanotechnology Division, ICAR-Central Inland Fisheries Research Institute, Barrackpore, Kolkata, West Bengal, 700120, India.
| |
Collapse
|
6
|
Palmeri S, Penco F, Bertoni A, Bustaffa M, Matucci-Cerinic C, Papa R, Drago E, Caorsi R, Corcione A, Bocca P, Scarone C, Rubartelli A, Volpi S, Gattorno M, Prigione I. Pyrin Inflammasome Activation Defines Colchicine-Responsive SURF Patients from FMF and Other Recurrent Fevers. J Clin Immunol 2024; 44:49. [PMID: 38231350 DOI: 10.1007/s10875-023-01649-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/20/2023] [Indexed: 01/18/2024]
Abstract
Syndrome of undifferentiated recurrent fever (SURF) is characterized by recurrent fevers, a lack of confirmed molecular diagnosis, and a complete or partial response to colchicine. Despite the clinical similarities to familial Mediterranean fever (FMF), the underlying inflammatory mechanisms of SURF are not yet understood. We here analyzed the in vitro activation of the pyrin inflammasome in a cohort of SURF patients compared to FMF and PFAPA patients. Peripheral blood mononuclear cells (PBMC) were collected from SURF (both colchicine-treated and untreated), FMF, PFAPA patients, and healthy donors. PBMC were stimulated ex vivo with Clostridium difficile toxin A (TcdA) and a PKC inhibitor (UCN-01), in the presence or absence of colchicine. The assembly of the pyrin inflammasome was evaluated by measuring the presence of apoptosis-associated Speck-like protein containing caspase recruitment domain (ASC) specks in monocytes using flow cytometry. IL-1β secretion was quantified using an ELISA assay. No differences in TcdA-induced activation of pyrin inflammasome were observed among FMF, PFAPA, and healthy donors. Untreated SURF patients showed a reduced response to TcdA, which was normalized after colchicine treatment. In contrast to FMF, SURF patients, similar to PFAPA patients and healthy donors, did not exhibit pyrin inflammasome activation in response to UCN-01-mediated pyrin dephosphorylation. These data demonstrate that in vitro functional analysis of pyrin inflammasome activation can differentiate SURF from FMF and PFAPA patients, suggesting the involvement of the pyrin inflammasome in the pathophysiology of SURF.
Collapse
Affiliation(s)
- Serena Palmeri
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Dipartimento Di Neuroscienze, Riabilitazione, OftalmologiaGenetica e Scienze Materno-Infantili (DINOGMI), Università Degli Studi Di Genova, Genoa, Italy
| | - Federica Penco
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Arinna Bertoni
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marta Bustaffa
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Riccardo Papa
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Enrico Drago
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Dipartimento Di Neuroscienze, Riabilitazione, OftalmologiaGenetica e Scienze Materno-Infantili (DINOGMI), Università Degli Studi Di Genova, Genoa, Italy
| | - Roberta Caorsi
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Anna Corcione
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Paola Bocca
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Cristina Scarone
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Stefano Volpi
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Dipartimento Di Neuroscienze, Riabilitazione, OftalmologiaGenetica e Scienze Materno-Infantili (DINOGMI), Università Degli Studi Di Genova, Genoa, Italy
| | - Marco Gattorno
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy.
| | - Ignazia Prigione
- UOC Reumatologia E Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
7
|
Cabral A, Cabral JE, Wang A, Zhang Y, Liang H, Nikbakht D, Corona L, Hoffman HM, McNulty R. Differential Binding of NLRP3 to non-oxidized and Ox-mtDNA mediates NLRP3 Inflammasome Activation. Commun Biol 2023; 6:578. [PMID: 37253813 PMCID: PMC10229695 DOI: 10.1038/s42003-023-04817-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 04/06/2023] [Indexed: 06/01/2023] Open
Abstract
The NLRP3 inflammasome is a key mediator of the innate immune response to sterile tissue injury and is involved in many chronic and acute diseases. Physically and chemically diverse agents activate the NLRP3 inflammasome. Here, we show that NLRP3 binds non-oxidized and Ox-mtDNA differentially, with a half maximum inhibitory concentration (IC50) for non-oxidized and Ox-mtDNA of 4 nM and 247.2 nM, respectively. The NLRP3 Neonatal-Onset Multisystem Inflammatory Disease (NOMIDFCAS) gain of function mutant could bind non-oxidized mtDNA but had higher affinity for Ox-mtDNA compared to WT with an IC50 of 8.1 nM. NLRP3 lacking the pyrin domain can bind both oxidized and non-oxidized mtDNA. Isolated pyrin domain prefers Ox-mtDNA. The NLRP3 pyrin domain shares a protein fold with DNA glycosylases and generate a model for DNA binding based on the structure and sequence alignment to Clostridium acetobutylicum and human OGG1, an inhibitor of Ox-mtDNA generation, 8-oxoguanine DNA glycosylases. We provide a new model for how NLRP3 interacts with Ox-mtDNA supported by DNA binding in the presence of a monoclonal antibody against the pyrin domain. These results give new insights into the mechanism of inflammasome assembly, and into the function of reactive oxygen species in establishing a robust immune response.
Collapse
Affiliation(s)
- Angela Cabral
- Department of Molecular Biology and Biochemistry, University of California Irvine, Steinhaus Hall, Irvine, CA, 92694-3900, USA
| | - Julia Elise Cabral
- Department of Molecular Biology and Biochemistry, University of California Irvine, Steinhaus Hall, Irvine, CA, 92694-3900, USA
| | - Angelina Wang
- Department of Molecular Biology and Biochemistry, University of California Irvine, Steinhaus Hall, Irvine, CA, 92694-3900, USA
| | - Yiyang Zhang
- Department of Molecular Biology and Biochemistry, University of California Irvine, Steinhaus Hall, Irvine, CA, 92694-3900, USA
| | - Hailin Liang
- Department of Molecular Biology and Biochemistry, University of California Irvine, Steinhaus Hall, Irvine, CA, 92694-3900, USA
| | - Donya Nikbakht
- Department of Molecular Biology and Biochemistry, University of California Irvine, Steinhaus Hall, Irvine, CA, 92694-3900, USA
| | - Leslie Corona
- Department of Molecular Biology and Biochemistry, University of California Irvine, Steinhaus Hall, Irvine, CA, 92694-3900, USA
| | - Hal M Hoffman
- Division of Pediatric Allergy, Immunology, and Rheumatology, Rady Children's Hospital of San Diego, University of California, San Diego, San Diego, CA, USA
| | - Reginald McNulty
- Department of Molecular Biology and Biochemistry, University of California Irvine, Steinhaus Hall, Irvine, CA, 92694-3900, USA.
| |
Collapse
|
8
|
Wang DN, Ni JJ, Li JH, Gao YQ, Ni FJ, Zhang ZZ, Fang JY, Lu J, Yao YF. Bacterial infection promotes tumorigenesis of colorectal cancer via regulating CDC42 acetylation. PLoS Pathog 2023; 19:e1011189. [PMID: 36812247 PMCID: PMC9987831 DOI: 10.1371/journal.ppat.1011189] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/06/2023] [Accepted: 02/07/2023] [Indexed: 02/24/2023] Open
Abstract
Increasing evidence highlights the role of bacteria in promoting tumorigenesis. The underlying mechanisms may be diverse and remain poorly understood. Here, we report that Salmonella infection leads to extensive de/acetylation changes in host cell proteins. The acetylation of mammalian cell division cycle 42 (CDC42), a member of the Rho family of GTPases involved in many crucial signaling pathways in cancer cells, is drastically reduced after bacterial infection. CDC42 is deacetylated by SIRT2 and acetylated by p300/CBP. Non-acetylated CDC42 at lysine 153 shows an impaired binding of its downstream effector PAK4 and an attenuated phosphorylation of p38 and JNK, consequently reduces cell apoptosis. The reduction in K153 acetylation also enhances the migration and invasion ability of colon cancer cells. The low level of K153 acetylation in patients with colorectal cancer (CRC) predicts a poor prognosis. Taken together, our findings suggest a new mechanism of bacterial infection-induced promotion of colorectal tumorigenesis by modulation of the CDC42-PAK axis through manipulation of CDC42 acetylation.
Collapse
Affiliation(s)
- Dan-Ni Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin-Jing Ni
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Hui Li
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ya-Qi Gao
- State Key Laboratory for Oncogenes and Related Genes; Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang-Jing Ni
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen-Zhen Zhang
- Department of Pathology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Yuan Fang
- State Key Laboratory for Oncogenes and Related Genes; Division of Gastroenterology and Hepatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Lu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (JL); (Y-FY)
| | - Yu-Feng Yao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Microbial Metabolism, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, China
- * E-mail: (JL); (Y-FY)
| |
Collapse
|
9
|
Torre C, Boyer L. Effector-Triggered Trained Immunity: An Innate Immune Memory to Microbial Virulence Factors? Toxins (Basel) 2022; 14:toxins14110798. [PMID: 36422972 PMCID: PMC9696518 DOI: 10.3390/toxins14110798] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/04/2022] [Accepted: 11/15/2022] [Indexed: 11/19/2022] Open
Abstract
In the last decade, a major dogma in the field of immunology has been called into question by the identification of a cell autonomous innate immune memory. This innate immune memory (also named trained immunity) was found to be mostly carried by innate immune cells and to be characterized by an exacerbated inflammatory response with a heightened expression of proinflammatory cytokines, including TNF-α, IL-6 and IL-1β. Unlike the vast majority of cytokines, IL-1β is produced as a proform (pro-IL-1β) and requires a proteolytic cleavage to exert its biological action. This cleavage takes place mainly within complex molecular platforms named inflammasomes. These platforms are assembled upon both the infectious or sterile activation of NOD-like receptors (NLRs), thereby allowing for the recruitment and activation of caspases and the subsequent maturation of pro-IL-1β into IL-1β. The NLRP3 inflammasome has recently been implicated both in western diet-induced trained immunity, and in the detection of microbial virulence factors (effector-triggered immunity (ETI)). Here, we will attempt to link these two immune processes and provide arguments to hypothesize the existence of trained immunity triggered by microbial virulence factors (effector-triggered trained immunity (ETTI)).
Collapse
|
10
|
Bacteria in tumors “hit the road” together. Cell 2022; 185:1292-1294. [DOI: 10.1016/j.cell.2022.03.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 11/20/2022]
|
11
|
Planès R, Santoni K, Meunier E. Analysis of Bacteria-Triggered Inflammasome: Activation in Neutrophils by Immunoblot. Methods Mol Biol 2022; 2523:265-279. [PMID: 35759203 DOI: 10.1007/978-1-0716-2449-4_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Detection of microbes relies on the expression of germline-encoded pattern recognition receptors (PRRs). While PRRs can directly sense conserved pattern expressed by various microbes, they can also induce effector-triggered immunity (ETI) by sensing pathogenic alterations of cellular homeostasis. One consequence of ETI is the death of the infected cell through the induction of inflammasome-dependent cell death, namely, pyroptosis. Such process can be easily studied in macrophages and epithelial cells, yet neutrophils encode an arsenal of proteolytic enzymes that imped easy and reliable study of ETI-triggered inflammasome response. Here, we describe an immunoblotting methodology to study both ETI- and PRR-driven inflammasome responses in neutrophils upon bacterial infections. This method is also transposable to other microbial pathogen- and toxin-induced inflammasome response in neutrophils.
Collapse
Affiliation(s)
- Rémi Planès
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Karin Santoni
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France
| | - Etienne Meunier
- Institute of Pharmacology and Structural Biology (IPBS), University of Toulouse, CNRS, Toulouse, France.
| |
Collapse
|